Language selection

Search

Patent 2605080 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2605080
(54) English Title: MATERIALS AND METHODS FOR ALTERING AN IMMUNE RESPONSE TO EXOGENOUS AND ENDOGENOUS IMMUNOGENS, INCLUDING SYNGENEIC AND NON-SYNGENEIC CELLS, TISSUES OR ORGANS
(54) French Title: MATERIAUX ET METHODES PERMETTANT DE MODIFIER UNE REPONSE IMMUNITAIRE A DES AGENTS IMMUNOGENES EXOGENES ET ENDOGENES, Y COMPRIS DES CELLULES, TISSUS OU ORGANES SYNGENEIQUES ET NON SYNGENEIQUES
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 5/071 (2010.01)
  • A61L 27/38 (2006.01)
  • A61P 37/06 (2006.01)
  • C12N 11/00 (2006.01)
  • A61K 35/12 (2006.01)
  • A61K 35/14 (2006.01)
(72) Inventors :
  • EDELMAN, ELAZER R. (United States of America)
  • NUGENT, HELEN MARIE (United States of America)
  • METHE, HEIKO (United States of America)
(73) Owners :
  • MASSACHUSETTS INSTITUTE OF TECHNOLOGY (United States of America)
  • SHIRE REGENERATIVE MEDICINE, INC. (Not Available)
(71) Applicants :
  • MASSACHUSETTS INSTITUTE OF TECHNOLOGY (United States of America)
  • PERVASIS THERAPEUTICS, INC. (United States of America)
(74) Agent: MBM INTELLECTUAL PROPERTY LAW LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2006-04-21
(87) Open to Public Inspection: 2006-11-02
Examination requested: 2009-06-03
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2006/015555
(87) International Publication Number: WO2006/116357
(85) National Entry: 2007-10-15

(30) Application Priority Data:
Application No. Country/Territory Date
60/673,417 United States of America 2005-04-21
60/682,217 United States of America 2005-05-18

Abstracts

English Abstract




Disclosed herein are materials and methods for modulating an immunologically
adverse response to an exogenous or endogenous immunogen, including a cell,
tissue, or organ associated immunogen. An implantable material comprising
cells, such as but not limited to endothelial cells, anchored or embedded in a
biocompatible matrix can modulate an adverse immune or inflammatory reaction
to exogenous or endogenous immunogens, including response to non-syngeneic or
syngeneic cells, tissues or organs, exogenous immunogens or stimuli, as well
as ameliorate an autoimmune condition. The implantable material can be
provided prior to, coincident with, or subsequent to occurrence of the immune
response or inflammatory reaction. The implantable material can induce
immunological acceptance in a transplant patient, reduce graft rejection and
reduce donor antigen immunogenicity.


French Abstract

L'invention concerne des matériaux et des méthodes permettant de moduler une réponse contraire au plan immunologique à un agent immunogène exogène ou endogène, y compris un agent immunogène associé à des cellules, tissus ou organes immunogènes. Un matériau implantable comprenant notamment, mais pas uniquement, des cellules endothéliales ancrées ou nichées dans une matrice biocompatible peut moduler une réaction immunitaire ou inflammatoire contraire à des agents immunogènes exogènes ou endogènes, notamment une réponse à des cellules, tissus ou organes non syngenéiques ou syngénéiques, à des agents ou des stimuli immunogènes exogènes, et améliorer un état auto-immun. Le matériau implantable peut être utilisé avant, pendant ou après la survenue d'une réponse immunitaire ou d'une réaction inflammatoire. Ce matériau implantable peut induire une acceptation immunologique chez un patient transplanté, réduire les risques de rejet de la greffe et atténuer l'immuno-génicité antigénique du donneur.

Claims

Note: Claims are shown in the official language in which they were submitted.




What we claim is:



1. A method of reducing an immune response or an inflammatory
reaction, comprising the step of:

providing to a recipient an implantable material comprising
a biocompatible matrix; and,

anchored or embedded endothelial cells, endothelial-like cells,
or analogs thereof,

wherein said implantable material is provided to said recipient in an
amount sufficient to reduce the immune response or inflammatory
reaction in said recipient.

2. The method of claim 1 wherein the providing step is prior to,
coincident with, or subsequent to administration to said recipient of one or
more doses of a cell, tissue or organ from a syngeneic or non-syngeneic
donor.

3. The method of claim 1 wherein the providing step is prior to,
coincident with, or subsequent to occurrence of the immune response or
inflammatory reaction.






4. A method of inducing acceptance in a patient, comprising the step of:
providing an implantable material comprising a biocompatible matrix and
anchored or embedded endothelial cells, endothelial-like cells, or analogs
thereof, prior to, coincident with, or subsequent to transplantation of
autograft, xenograft or allograft cells, tissue or organ in said patient in an

amount effective to induce acceptance in said patient.

5. A method of reducing donor antigen immunogenicity, comprising the
step of:

providing an implantable material comprising a biocompatible matrix and
anchored or embedded endothelial cells, endothelial-like cells, or analogs
thereof prior to, coincident with, or subsequent to introduction of said donor

antigen to a recipient in an amount effective to reduce donor antigen
immunogenicity.

6. The method of claim 1, 4 or 5 wherein said providing step occurs
prior to, coincident with, or subsequent to administration to said recipient
of
an immunosuppressive agent.

7. The method of claim 6 wherein said immunosuppressive agent
resides in said implantable material during coincident administration.

8. The method of claim 5 wherein said donor and recipient are the same.
9. The method of claim 1, 4, 5 or 8 wherein said recipient has an
autoimmune disease.

10. An implantable material suitable for use with any one of claims 1-9.



91



11. The implantable material of claim 10 wherein the endothelial-like
cells or analogs are non-endothelial cells.

12. The implantable material of claim 10 wherein the cells are autogenic,
allogenic, xenogenic or genetically-modified variants of any one of the
foregoing.

13. The implantable material of claim 10 wherein the cells are vascular
endothelial cells.

14. A method of transplanting to a recipient a syngeneic or non-
syngeneic cell, tissue or organ transplant, comprising the step of:
providing to said recipient an implantable material comprising a
biocompatible matrix and anchored or embedded endothelial cells,
endothelial-like cells, or analogs thereof, prior to, coincident with, or
subsequent to transplantation such that said transplanted syngeneic or
non-syngeneic cell, tissue or organ is not rejected by said recipient.

15. The method of claim 4 or 14 wherein said transplanted cell, tissue or
organ comprises non-endothelial cells.

16. The method of claim 5 wherein said donor antigen comprises a non-
endothelial cell antigen.

17. The method of claim 14 further comprising the step of administering
an immunosuppresive agent prior to, coincident with, or subsequent to
transplantation.

18. A cell suitable for use with the implantable material of any one of
claims 1-17.



92



19. The cell of claim 18 wherein said endothelial-like cell or its analog is
a non-endothelial cell.

20. The cell of claim 18 wherein said analog is non-natural.

21. The cell of claim 18 wherein said cell, when anchored to or
embedded within a biocompatible matrix, reduces a recipient's humoral or
cellular immune response to a non-syngeneic donor cell, tissue or organ.
22. The cell of claim 18 wherein said cell, when anchored to or
embedded within a biocompatible matrix, exhibits diminished
immunogenicity.

23. The cell of claim 22 wherein said diminished immunogenicity is
reduced expression of MHC or reduced capacity to bind, activate or promote
maturation of innate immune cells, when anchored to or embedded within a
biocompatible matrix, wherein said innate immune cells are selected from
the group consisting of NK cells, dendritic, cells, monocytes, and
macrophages.

24. The cell of claim 18 wherein said cell, when anchored to or
embedded within a biocompatible matrix, exhibits reduced expression of
MHC, costimulatory molecules or adhesion molecules.



93



25. The cell of claim 18 wherein said cell, when anchored to or
embedded within a biocompatible matrix and co-cultured with a dendritic
cell, inhibits expression by said dendritic cell of HLA-DR, IL12, Toll-like
receptor or CD83; promotes uptake of dextran by said dendritic cell; or
blocks dendritic cell-induced lymphocyte proliferation; or when co-cultured
with adaptive immune cells inhibits proliferation, activation or
differentiation
of said cells, wherein adaptive immune cells are selected from the group
consisting of B-lymphocytes and T-lymphocytes.

26. An implantable material comprising a biocompatible matrix and the
anchored or embedded endothelial cell, endothelial-like cell, or analog
thereof of claim 22.

27. An implantable material comprising a biocompatible matrix and the
anchored or embedded endothelial cell, endothelial-like cell, or analog
thereof of claim 24.

28. An implantable material comprising a biocompatible matrix and the
anchored or embedded endothelial cell, endothelial-like cell, or analog
thereof of claim 25.

29. A cell bank comprising the cell of claim 18 or any one of claims 23-
25.

30. A bank of implantable material comprising the implantable material
of claim 10.



94



31. A bank of implantable material, wherein said implantable material
comprises a biocompatible matrix and the anchored or embedded endothelial
cell, endothelial-like cell, or analog thereof of claim 22.

32. A bank of implantable material, wherein said implantable material
comprises a biocompatible matrix and the anchored or embedded endothelial
cell, endothelial-like cell, or analog thereof of claim 24.

33. A bank of implantable material, wherein said implantable material
comprises a biocompatible matrix and the anchored or embedded endothelial
cell, endothelial-like cell, or analog thereof of claim 25.

34. The implantable material of claim 10 wherein said implantable
material is a solid or non-solid.

35. The implantable material of claim 10 wherein said implantable
material is provided to the recipient by implantation, injection or infusion.
36. An implantable material for reducing an immune response to a non-
syngeneic cell, tissue or organ, wherein said implantable material comprises:

a biocompatible matrix; and, anchored thereto or embedded therein,
endothelial cells, endothelial-like cells, or analogs thereof;

or
tissue, or organ, or a segment thereof;

wherein an effective amount of said implantable material reduces the
recipient's immune response to said non-syngeneic cell, tissue or
organ.






37. The implantable material of claim 36 wherein said non-syngeneic
cell, tissue or organ is that of the recipient suffering from an autoimmune
disease.

38. A method of reducing an immune response or an inflammatory
reaction resulting from exposure to an exogenous immunogen, comprising
the step of:

providing to a recipient an implantable material comprising
a biocompatible matrix; and,

anchored or embedded endothelial cells, endothelial-like cells,
or analogs thereof,

wherein said implantable material is provided to said recipient in an
amount sufficient to reduce the immune response or inflammatory
reaction in said recipient resulting from exposure to said exogenous
immunogen.

39. The method of claim 38 wherein the providing step is prior to,
coincident with, or subsequent to occurrence of the immune response or
inflammatory reaction.

40. The method of claim 38 wherein said exogenous immunogen is
naturally occurring.

41. The method of claim 38 wherein said exogenous immunogen is
selected from the group consisting of pharmaceutical agents, toxins, surgical
implants, infectious agents and chemicals.



96



42. The method of claim 38 wherein said exogenous immunogen is an
exogenous stimulus selected from the group consisting of environmental
stress, injury and exposure.



97

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
MATERIALS AND METHODS FOR ALTERING AN IMMUNE RESPONSE
TO EXOGENOUS AND ENDOGENOUS IMMUNOGENS, INCLUDING
SYNGENEIC AND NON-SYNGENEIC CELLS, TISSUES OR ORGANS

Field of the Invention

This invention is directed to materials and methods for modulating an
immunologically adverse response to an exogenous or endogenous immunogen,
including a cell-, tissue-, or organ-associated iminunogen. For example, the
present
invention can modulate an adverse immune response or inflammatory reaction to

exogenous or endogenous immunogens, including non-syngeneic or syngeneic
cells,
tissues or organs, as well as ameliorate an autoimmune condition.

Background of the Invention

Research on xenotransplantation has been intensified over the past years to
alleviate organ shortage. However, host immune responses present a formidable
barrier to transplantation across species. Whereas natural antibodies cause

immediate rejection of such discordant transplants, endothelial cell (EC)
injury and
activation of graft vessel lining EC play a pivotal role in initiating chronic
graft
rejection. Disruption of the integrity of the endothelial layer is of
undoubted
importance in numerous conditions, including syngeneic and non-syngeneic
tissue

transplants as well as infectious, neoplastic, inflammatory and cardiovascular
diseases.

1


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Heretofore immunomodulation and transplant acceptance have required
reliance on systemically-administered immunosuppressive agents. While such
agents permit some degree of transplant acceptance, success is limited and
perhaps
of more significance, a patient's immune system is thoroughly compromised as a

result of such agents. Thus a need still remains for therapeutic materials and
treatment paradigms which can achieve immunomodulation absent the toxicity and
adverse affects on a patient's immune system.

Similarly, exogenous immunogens or stimuli have posed a clinical challenge.
These, too, can result in adverse immunological events or inflammatory
reactions
which necessitate treatment. Heretofore, clinical management of such adverse

events has relied almost exclusively on treatments with pharmaceutical agents
which
suppress the immune system non-specifically. - I
Autoimmune diseases and other similar diseases are yet another clinical

manifestation of heightened inflammatory reactions or adverse immune
responses.
Successful management of such diseases has eluded clinicians to date.

An object of the present invention is to provide a tissue engineering solution
for achieving iminunomodulation without reliance on chemicals or
pharmaceuticals
which compromise a patient's immune system. This tissue engineering solution
can
be employed to alter, in a clinically practical manner, an immune response to

exogenous and endogenous immunogens, including non-syngeneic as well as
syngeneic cell-, tissue- or organ-associated immunogens. Another object of the
present invention is to facilitate a patient's acceptance of non-syngeneic as
well as
syngeneic cells, tissues or organs. Another object of the present invention is
to
employ this tissue engineering solution to modulate an inflammatory reaction
such

2


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
as that associated with injury and various diseases. Another object is to
utilize the
materials and methods of the present invention to manage autoimmunity and
related
diseases.

Summary of the Invention

The present invention exploits the discovery that cells anchored to and/or
embedded within a biocompatible matrix, preferably one having a three-
dimensional
configuration, can modulate an immunologically adverse response or
inflammatory
reaction to any exogenous or endogenous immunogen. Immunogen includes any
syngeneic or non-syngeneic immunogen, including a cell-, tissue-, or organ-

i.o associated immunogen, as well as injury, disease and environmental
stimuli.
In one aspect, the present invention is a method of reducing an immune
response or an inflammatory reaction. According to this method, a recipient is
provided an implantable material coinprising a biocompatible matrix and
anchored
and/or embedded endothelial cells, endothelial-like cells, or analogs thereof.
The

implantable material is provided to the recipient in an amount sufficient to
reduce
the immune response or inflammatory reaction in the recipient.

According to the invention, the providing step can occur prior to, coincident
with, or subsequent to administration to the recipient of one or more doses of
a cell,
tissue or organ from a syngeneic or non-syngeneic donor. According to another

embodiment, the providing step is prior to, coincident with, or subsequent to
occurrence of an immune response or inflammatory reaction. According to
another
embodiment, the method reduces an immune response or an inflammatory response
by modulating immunological memory.

3


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
In a related aspect, the present invention is a method of inducing
immunological acceptance in a patient. According to this method, the patient
is
provided an implantable material comprising a biocompatible matrix and
anchored
and/or embedded endothelial cells, endothelial-like cells, or analogs thereof,
prior

to, coincident with, or subsequent to transplantation of autograft, xenograft
or
allograft cells, tissue or organ in an amount effective to induce acceptance
in the
patient.

Additionally, the present invention is directed to a method of reducing donor
antigen immunogenicity. According to this method, an implantable material

comprising a biocompatible matrix and anchored and/or embedded endothelial
cells,
endothelial-like cells, or analogs thereof are presented prior to, coincident
with, or
subsequent to introduction of the donor antigen to a recipient in an amount
effective
to reduce donor antigen iinmunogenicity. According to another embodiment, the
donor and recipient are the same. According to a further embodiment, the
recipient

has an autoimmune disease. According to yet another embodiment, the donor
antigen comprises a non-endothelial cell antigen.

According to various other embodiments, the providing step occurs prior to,
coincident with, or subsequent to administration to the recipient of an
immunosuppressive agent. The immunosuppressive agent can reside in the

iinplantable material.

Moreover, the present invention is also directed to a method of transplanting
to a recipient a syngeneic or non-syngeneic cell, tissue or organ transplant.
According to this method, a recipient is provided an implantable material
comprising a biocompatible matrix and anchored and/or embedded endothelial
cells,

4


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
endothelial-like cells, or analogs thereof, prior to, coincident with, or
subsequent to
transplantation such that the transplanted syngeneic or non-syngeneic cell,
tissue or
organ is not rejected by the recipient. According to one embodiment of the
method,
the transplanted cell, tissue or organ comprises non-endothelial cells.

In another aspect, the present invention is an implantable material
comprising a biocompatible matrix and cells anchored thereto and/or embedded
tlierein. According to one currently preferred embodiment, the cells are
endothelial
cells, endothelial-like cells and/or analogs of either. In certain other
embodiments,
endothelial-like cells or analogs of the implantable material are non-
endothelial

cells. According to another embodiment, the cells of the implantable material
are
autogenic, allogenic, xenogenic or genetically-modified variants of any one of
the
foregoing cell types. According to a further preferred embodiment, the cells
of the
implantable material are vascular endothelial cells. According to one
embodiment,
the implantable material is a solid or non-solid. According to yet another,
the

implantable material is provided to the recipient by implantation, injection
or
infusion.

The present invention is also directed to an implantable material for reducing
an immune response to a syngeneic or non-syngeneic cell, tissue or organ.
According to this aspect of the invention, the implantable material comprises
a

biocompatible matrix and, anchored thereto and/or embedded therein,
endothelial
cells, endothelial-like cells, or analogs thereof. According to this aspect of
the
invention, an effective amount of the implantable material reduces the
recipient's
immune response to the syngeneic or non-syngeneic cell, tissue or organ.

5


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
According to one embodiment of this aspect of the present invention, the cell,
tissue
or organ is that of the recipient suffering from an autoimmune disease.

The invention is also directed to a variation of the above-described
implantable material which is useful for reducing an immune response to a non-

syngeneic cell, tissue or organ, wlierein said iinplantable material comprises
cells,
tissue, or organ or a segment thereof anchored to and/or embedded within the
biocompatible matrix. An effective amount of this implantable material reduces
the
recipient's immune response to a non-syngeneic cell, tissue or organ. The non-
syngeneic cell, tissue or organ is that of the recipient suffering from an
autoimmune
io disease.

In a furtlier aspect, the present invention is a cell suitable for use with
the
implantable material of any one of inventions described herein. According to
one
embodiment, the endothelial-like cell or its analog is a non-endothelial cell.

According to another embodiment, the analog is non-natural. According to a
further
embodiment, the cell, when anchored to and/or embedded within a biocompatible
matrix, reduces a recipient's huinoral or cellular immune response to a
syngeneic or
non-syngeneic donor cell, tissue or organ.

According to another embodiment, the cell, when anchored to and/or
einbedded within a biocompatible matrix, exhibits diminished immunogenicity.
According to one embodiment, the cell exhibits diminished immunogenicity by

exhibiting reduced expression of MHC or reduced capacity to bind, activate or
promote maturation of innate immune cells, when anchored to and/or embedded
within a biocompatible matrix, wherein said innate immune cells are selected
from
the group consisting of NK cells, dendritic, cells, monocytes, and
macrophages.

6


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
According to another embodiment, the cell, when anchored to and/or
embedded within a biocompatible matrix, exhibits reduced expression of
costimulatory molecules or adhesion molecules. According to a further
embodiment, the cell, when anchored to and/or embedded within a biocompatible

matrix and co-cultured with a dendritic cell, inhibits expression by said
dendritic cell
of HLA-DR, IL12, Toll-like receptor or CD83; promotes uptake of dextran by
said
dendritic cell; or blocks dendritic cell-induced lymphocyte proliferation; or
when
co-cultured with adaptive immune cells inhibits proliferation, activation or

differentiation of said cells, wherein adaptive immune cells are selected from
the
group consisting of B-lymphocytes and T-lymphocytes.

In another aspect, the present invention is a cell bank comprising any one of
the cells described herein. In a furtlier aspect, the present invention is a
bank
coinprising any one of the implantable materials described herein.

In a further aspect, the present invention is a method of reducing an immune
response or an inflammatory reaction resulting from exposure to an exogenous
immunogen. According to this method, a recipient is provided with an
implantable
material comprising a biocompatible matrix and anchored or embedded
endothelial
cells, endothelial-like cells, or analogs thereof. The implantable material is
provided
to the recipient in an amount sufficient to reduce the immune response or

inflammatory reaction in the recipient resulting from exposure to the
exogenous
immunogen.

According to one embodiment of this method, the providing step is prior to,
coincident with, or subsequent to occurrence of the immune response or
inflammatory reaction. According to another embodiment, the exogenous

7


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
immunogen is naturally occurring. According to a further embodiment, the
exogenous immunogen is selected from the group consisting of pharmaceutical
agents, toxins, surgical implants, infectious agents and chemicals. According
to
another embodiment, the exogenous immunogen is an exogenous stimulus selected

from the group consisting of environmental stress, injury and exposure.
Brief Description of the Drawings

Figures 1A, 1B and 1C graphically depict levels of circulating PAE-specific
antibodies according to an illustrative embodiment of the invention.

Figure 2 graphically depicts lytic activity of splenocytes according to an
illustrative embodiment of the invention.

Figure 3A graphically depicts the frequencies of cytokine-producing cells
according to an illustrative embodiment of the invention.

Figure 3B depicts representative ELISPOT wells according to an illustrative
embodiment of the invention.

Figure 3C graphically depicts the frequencies of T cells according to an
illustrative embodiment of the invention.

Figures 4A and 4B graphically plot levels of effector cells according to an
illustrative embodiment of the invention.

Figures 5A, 5B and 5C graphically depict antibody levels according to an
illustrative embodiment of the invention.

Figure 6 graphically depicts levels of splenocytes according to an
illustrative
embodiment of the invention.

8


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Figures 7A and 7B graphically depict antibody levels according to an
illustrative embodiment of the invention.

Figures 8A and 8B graphically depict the frequency of cytokine-producing
cells according to an illustrative embodiment of the invention.

Figures 9A and 9B graphically depict effector cell levels according to an
illustrative embodiment of the invention.

Figures 1 OA and 1 OB depict correlations between the frequency of Th2-
cytokine producing splenocytes and the extent of T cell differentiation into
effector
cells according to an illustrative embodiment of the invention.

Figure 11 graphically depicts the degree of damage to endothelial cells
according to an illustrative embodiment of the invention.

Figures which refer to "embedded" or "matrix-embedded" PAE, HAE or EC
mean matrix-anchored and/or matrix-embedded PAE, HAE, EC.

Detailed Description of the Invention

Tissue engineering is a promising approach to exploit endothelial cells,
endothelial-like cells, or analogs of either as a cellular therapy for
diseases
accompanied by or typified by adverse immunological components. For example,
certain diseases such as but not limited to vascular diseases provoke adverse
immunological responses and/or inflammatory reactions. The present invention
is

based on the discovery that cells such as endothelial cells which are anchored
to or
embedded in three-dimensional matrices, secrete essential regulatory factors
which
can ameliorate or otherwise modulate an adverse immunological response.

9


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
The implantable material of the present invention was developed on the
principals of tissue engineering and represents a novel approach to addressing
the
herein-described clinical needs. The implantable material of the present
invention is
unique in that the viable cells anchored to and/or embedded within the

biocompatible matrix are able to supply to the site of administration multiple
cell-
based products in physiological proportions under physiological feed-back
control.
As described elsewhere herein, the cells suitable for use with the implantable
material are endothelial, endothelial-like cells, or analogs of each of the
foregoing.
Local delivery of multiple compounds by these cells and physiologically-
dynamic

lo dosing provide more effective regulation of the processes responsible for
modulating
an immune response. The implantable material of the present invention can
provide
an environment which mimics supportive physiology and is conducive to
modulation of an immune response.

This is an unexpected discovery since endothelial cells can play a pivotal
role in initiation of adverse allo- and xeno-immune responses. Moreover,
endothelial cells can activate T-cells tlirough antigen-mediated processes and
T-cell
activation can modify crucial endothelial cell function, including antigen
presentation via activation by cytokines, thereby contributing to an adverse
immune
response. And, endothelial cells constitutively express class I Major

Histocompatibility Complex (MHC) molecules, and IFN-y can induce endotlielial
cells to express class II MHC molecules which allows them to provide antigen-
dependent signals to CD8+ and CD4+ T-cells through the direct pathway.
Endothelial cells also can primarily provide costimulation to T-cells. In
addition,
the capacity to capture T-cells via endothelial expression of adhesion
molecules



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
allows formation of contact regions which furthers the adverse immune response
in
the form of inflammation. Furthermore, autoimmunity can exacerbate vascular
disease, in particular in the form of anti-endothelial cell antibodies. The
heightened
morbidity of cardiovascular diseases in concert with diabetes mellitus,
hypertension

and other disease states reflects the increased presence and potentcy of these
antibodies.

In contrast, as disclosed herein, matrix-anchored and/or -embedded
endothelial cells, when implanted in a host, act as powerful regulators of the
immune system as indicated by significant reduction in the expected systemic

immune response and/or local inflammation. As exemplified herein, the ability
of
such cells to ameliorate or modulate immune responsiveness has been
demonstrated
by comparing the immune response against free versus matrix-anchored and/or -
embedded endothelial cells in naive mice as well as mice with heightened
endothelial cell immune reactivity. Matrix-associated endothelial cells as
described

herein provide immune protection at multiple levels; human and porcine
endothelial
cells demonstrate a marked reduction in elaborated MHC class molecules;
costimulatory molecules; and adhesion molecules when matrix-anchored and/or-
embedded as disclosed herein.

Matrix anchoring and/or embedding of endothelial cells can also influence
formation of immunological memory as exemplified herein. Whereas
reimplantation of free, saline-suspended endothelial cell pellets alone or as
pellets
situated adjacent to an empty matrix evoked a significant increased humoral
and
cellular xenoresponse, rechallenging mice with matrix-anchored and/or -
embedded
endothelial cells led to a reduced lytic ability of splenocytes without
enhancing the

11


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
humoral immune responses. Moreover, a modest shift in the Thl/Th2 balance
towards the former was obvious in mice receiving matrix-anchored and/or -
embedded xenogeneic endothelial cells.

Thus, introduction of free endothelial cells adjacent to an empty matrix
failed
to reduce the host immune response indicating the importance of matrix-
anchoring
and/or - embedding. Failure of anchored and/or embedded endothelial cells to
express MHC II, costimulatory, and adhesion molecules upon stimulation could
account for the attenuated differentiation of T-cells in effector cells in
response to
implanted matrix-anchored and/or - embedded xenogeneic endothelial cells. As

explained herein, activation of mice splenocytes is inuted when exposed to
matrix-
anchored and/or - embedded xenogeneic endothelial cells in a MHC class II
dependent manner.

Overall the isotropic nature of endothelial cells contributes to this unique
form of immunomodulation wherein cell anchoring and/or embedding in a suitable
matrix provides immunoprotection through isolation or masking of critical
antigens.

It is well recognized that in vivo endothelial cell function is anchorage- and
density-
dependent. Previous studies have shown that the endotlielial basement membrane
(EBM) controls aspects of cell adhesion, spreading, migration, contractility,
differentiation, proliferation, protein synthesis and secretion. Furthermore,
EBM is

altered in many in vivo disease states, from diabetes to glomerulopathy to
atherosclerosis. Dysfunction of endothelial cells correlates with changes in
basement membrane composition cumulating in the degree of attachment of
endothelial cells, and the quality of basement membrane anchoring plays a role
for

endothelial cells immunobiology.

12


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
The present invention is based on the unexpected discovery that anchoring
and/or embedding endothelial cells in a suitable biocompatible matrix, such as
but
not limited to a 3-dimensional collagen-based matrix, can transform xenogeneic
endothelial cells into an immunologically non-offending cell phenotype. Such a

discovery can now be exploited by the skilled practitioner, following the
guidance
provided herein, as a tolerance-inducing approach to syngeneic or non-
syngeneic
therapies such as but not limited to allotransplantation or
xenotransplantation as
exemplified herein. For example, in a preferred embodiment of the present
invention, a clinician can diminish and/or delay rejection by implanting
matrix-

i.o anchored and/or - embedded endothelial cells prior to transplantation of
an allo- or
xenograft tissue or organ. For purposes of the present invention, blood is a
type of
tissue. Pre-treatment acclimates the recipient's immune system and can result
in a
reduced, attenuated and/or delayed immune response to a graft. The present

invention does not require that the implantable material comprise anchored-
and/or
embedded cells which are the same as or similar to those ultimately
transplanted in
the recipient. All that is required is that the implantable material
comprising

anchored- and/or embedded cells has an immunomodulatory effect when provided
to a recipient. In certain circumstances, a single administration prior to or

coincident with a transplant can be sufficient. In other circumstances,
multiple or
serial administrations are preferred. The skilled clinician will recognize
such
circumstances.

13


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
As is well recognized, even transplantation of allogeneic cells is often
accompanied by an immune response. A question of much interest is whether this
is
a constitutive and immutable property of foreign cells or one that can be
regulated.
The experiments set forth herein demonstrate that the immunogenicity of cells
that

are normally anchored to basement membranes can be markedly reduced if
implanted in a matrix-ancliored and/or - embedded state an effect not seen
when
these same cells were injected in a free state. Other experiments set forth
herein
investigate the influence of heightened anti-endothelial cell immunity which
is a
common clinical feature in a variety of autoimmune and endocrinological
diseases.

Additionally, certain of the experiments summarized herein demonstrate that
serial injections of free porcine aortic endothelial cells (PAE) induced
circulating
anti-PAE antibodies, elevating immunosensitivity. The response to subsequent
PAE
injections was even greater than that observed upon first exposure. In
contrast,
when PAE were implanted in a matrix-anchored and/or - embedded state, the

immune response to subsequent exposures was muted and dropped significantly
over time. Also, as illustrated below, the initial response to endothelial
cells is IgM-
mediated, lower than the subsequent IgG response and muted when preceded by
serial injections. The IgM response is more evident in naive than pre-
sensitized
animals and takes longer to abate after free PAE exposure to than after
exposure to

matrix-anchored and/or - embedded endothelial cells.

Pre-sensitization of mice with suspensions of free PAE resembles the IgGI-
driven anti-endothelial immunity seen in diabetes mellitus, hypertension and
autoimmune diseases. The cellular immune response to free and matrix-anchored
and/or - embedded cells followed the pattern of humoral immunity. Repeated

14


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
exposure to antigens resulted in increased formation of memory and
subsequently in
a more vigorous immune reaction by effector T cells. Hence, the induction of
xenoreactive IL-4- and IL-10-producing splenocytes and effector T cells was
elevated over time and visible after implantation of free endothelial cells in
naive

and pre-sensitized mice. In all mice, cytokine levels correlated linearly and
precisely with effector T cell induction further supporting the notion of a
Th2-driven
cellular response in xenoreactivity and accentuating the immunosilencing
aspects of
matrix-embedded endothelial cells to activate adaptive immune mechanisms.

Damage to implanted endothelial cells correlated with the extent of the immune
response elicited. Implanted cells were most profoundly affected after pre-
sensitization and with free PAE. The decreased induction of humoral and
cellular
immune responses in naive mice receiving matrix-embedded endothelial cells
resulted in a lesser degree of damage by host immune cells.

These experiments provide insights into the activation of and damage to

endothelial cells, suggesting a pivotal role for cell-matrix contact. The
honeycomb-
like structure of a currently preferred matrix, Gelfoam, allows endotlielial
cells to
associate with, or anchor to, or embed within its three-dimensional
configuration
and in certain embodiments, line the internal surfaces of this matrix in a
fashion
which simulates the appearance of confluent endothelium in quiescent vessels.
Thus

in certain embodiments, anchoring to and/or embedding endothelial cells within
a
matrix with the properties of Gelfoam resembles the physiologic three-
dimensional
state of intact endothelium. The experiments set forth below demonstrate that
matrix-anchoring and/or embedding not only protects endothelial cells from
host



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
immune reactions but changes the host's perception of endothelial cell
immunogenicity.

Thus, during disease for example, phenotypic transformation of endothelial
cells dislocated from an intact, matrix-adherent endogenous state to a free
state is
likely critical to initiation and perpetuation of vascular disease, for
example. The

teachings herein indicate that endothelial cell detachment precedes expression
of
adhesion, costiinulatory and MHC molecules which is then followed by
attraction of
immune cells, perpetuating endothelial activation and cell dainage. In this
regard,
the immunobiological and immunoreactive qualities of endothelial cells
correlate

with morphology and function. Endothelial cells from different vascular beds
and
divergent basement membrane connectivity demonstrate marked differences in
constitutive and inducible expression of adhesion, costimulatory and MHC-
molecules. Further, there is growing appreciation that deposition of
transitional
extracellular matrix proteins such as fibronectin and fibrinogen into the

subendothelial matrix as well as detachment of endothelial cells from the
basement
membrane affects intra-endothelial cell signaling.

As contemplated by the present invention, manipulation of cell phenotype,
immunogenicity, and function can be used to tailor the properties of tissue
engineered constructs developed in vitro for regenerative purposes; in
particular,

such a use of the present invention is clinically beneficial since current
cell-based
therapies are limited by profound host immune reactions. For example, the
present
invention is particularly useful for treatment of atherosclerotic disease
since the
presence of activated immune cells and inflammation are key pathophysiologic
components. Similarly, heightened anti-endothelial immunity has been
identified as

16


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
a pivotal rate-limiting effect for endothelial cell-based therapies, such as
but not
limited therapies involving seeding of the interior of a vascular structure
with cells
or tissue. In contrast, the present invention can be exploited to manage
endothelial
cell phenotypic shifts which occur in vascular patliology, e.g., via
dearrangement of

cell-matrix contact, and appropriately targeted therapeutic options can then
be
implemented in the clinic using the materials and methods of the present
invention.
Taken together, the teachings presented herein also demonstrate that features
of a matrix such as but not limited to biocompatability, porosity, three-

dimensionality, can support the growth of a population of endothelial cells
and can
lo modulate the immunogenicity of such cells. Endothelial cells anchored to
and/or
embedded within a three-dimensional matrix elicited far less activation of
host
immune mechanisms and were subject to far lower attack and damage from host
immune cells. Findings in naive mice were amplified in hosts with heightened
anti-
endothelial immunity. In vivo studies presented herein show a marked decrease
in

the Th2-driven immune response in animals implanted with a matrix such as
Gelfoam comprising anchored and/or embedded endothelial cells versus animals
injected with free endothelial cells. In order for endothelial cells to
activate naive
host T-cells, two signals are required: 1) antigen-presentation in the context
of MHC
molecules expressed on the donor endothelial cells and 2) a second signal
provided

2o by a costimulatory molecule also expressed on the donor endothelial cell
surface.
Therefore, while not wishing to be bound by theory, one possible explanation
for the
observed results is that the interaction between a biocompatible matrix and
embedded endothelial cells results in a decrease in surface expression of
crucial
costimulatory, MHC and/or adhesion molecules on the donor endothelial cells.

17


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Indeed, in vitro analysis of critical adhesion, MHC-II and co-stimulatory
molecule
expression on both PAE and HAE (human aortic endothelial cells) show a matrix-
anchored and/or embedded dependent profile. The expression profiles of
adhesion
(E-selectin, P-selectin, ICAM-1, VCAM-1, and CD58), costimulatory (CD40,

CD80, CD86) and MHC-II molecules were all reduced in endotlielial cells
anchored
to and/or embedded with a Gelfoam matrix as compared to the same endothelial
cells grown on standard tissue culture plates. P-selectin, E-selectin and VCAM-
1
are closely associated with T-cell recruitment at sites of immune
inflammation.
Because antigen presentation to CD4+ T-cells via MHC class II molecules is

io essential for host immune recognition in the setting of non-vascularized
xenogeneic
implants, the observed reduced MHC-II expression on matrix-anchored and/or -
embedded endothelial cells translated into a reduced proliferative response of
host
splenocytes. Furthermore, repeated in vitro exposure of the same splenocytes
to
endothelial cells grown on tissue culture plates elicited a more vigorous
secondary

response, whereas there was no increased secondary response and therefore no
memory of prior exposure to matrix-anchored and/or - embedded endothelial
cells.
These in vitro findings correlate with the significantly muted immune reaction
observed in rats and mice after implantation and re-challenge with matrix-
anchored
and or embedded endothelial cells as exemplified herein.

Similarly, a mechanism by which culturing endothelial cells in a
biocompatible matrix such as but not limited to Gelfoam affects expression of
MHC
class II molecules, and subsequent endothelial immunogenicity in vitro, was
further
elucidated by investigating intracellular signaling pathways. Endothelial
expression
of MHC class II molecules is induced by proinflammatory cytokines (e.g.
interferon
18


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
(IFN)-y) that are secreted by activated immune cells (e.g. T-cells). Binding
of
proinflammatory cytokines to their receptors on endothelial cells initiates an
intracellular signaling cascade resulting in phosphorylation of Janus protein
tyrosine
kinase (e.g. JAK-1 and 2) and signal transducer and activators of
transcription (e.g.

STAT-1). Activation of JAK and STAT are usually tightly regulated within a
target
cell. As set forth below, detailed in vitro analyses demonstrated differences
in IFN-
y induced intracellular signaling pathways between endothelial cells grown to
confluence on tissue culture plates as compared to those anchored to and/or
embedded or within Gelfoam matrices. Gelfoam-embedded HAE exhibited lower

io rates of STAT-phosphorylation and activation of the crucial interferon-
regulatory
factor-1 (IRF- 1) with no change in surface IFN-y receptor expression. Lower
rates
of JAK activation were also seen upon stimulation of HAE in Gelfoam with IFN-
y.

Upon further investigation, it was observed that non-IFN-y stimulated HAE
grown on a Gelfoam matrix expressed significantly higher levels of the

counteracting inhibitory molecule, Suppressor of Cytokine Signaling (SOCS)-1
and
3, than HAE grown on tissue culture plates. One explanation therefore for the
muted IFN-y induced intracellular signaling in Gelfoam-embedded HAE is that
the
increased levels of SOCS-1 and 3 resulted in an increase in the threshold for
cytokine-induced activation of endothelial cells.

In a currently preferred embodiment, the implantable material of the present
invention comprising anchored and/or embedded endothelial cells is implanted
at
any non-luminal site. Thus, immediate exposure of the donor cells to the host
circulation is not required. Recent evidence has demonstrated the importance
of the
soluble endothelial factor CX3CL1 (fractalkine) for attraction of immune cells
(i.e.,

19


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
natural killer cells) and surface expressed forms of fractalkine for adherence
of those
immune cells. Given that only modest cellular infiltration in and around
implantation sites of xeno- and allogeneic matrix-anchored and/or embedded
endothelial cells was observed, release of soluble and surface expression of

fractalkine on HAE was quantified. As illustrated in experiments set forth
below,
matrix-anchored and/or embedded endothelial cells showed reduced secretion and
down-regulation of fractalkine surface expression upon cytokine stimulation as
compared to HAE grown on tissue culture plates. This resulted in significantly
less
adherence of human natural killer cells to matrix-anchored and/or embedded HAE
in.
vitro.

Taken together, the changes in intracellular signaling, increased levels of
SOCS-1 and 3 (resulting in attenuated expression of MHC-II molecules and
subsequent T-cell activation) as well as reduced secretion and surface
expression of
fractalkine in matrix-anchored and/or embedded endothelial cells as compared
to

cells grown on tissue culture plates indicated an altered endothelial cell
immunogenicity attributable to matrix-embedding.

Cell Source. As described herein, the implantable material of the present
invention comprises cells which can be syngeneic, allogeneic, xenogeneic or
autologous. In certain embodiments, a source of living cells can be derived
from a

t20 suitable donor. In certain other embodiments, a source of cells can be
derived from
a cadaver or from a cell bank.

In one currently preferred embodiment, cells are endothelial cells. In a
particularly preferred embodiment, such endothelial cells are obtained from
vascular
tissue, preferably but not limited to arterial tissue. As exemplified below,
oiie type



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
of vascular endothelial cell suitable for use is an aortic endothelial cell.
Another
type of vascular endothelial cell suitable for use is umbilical cord vein
endothelial
cells. And, another type of vascular endothelial cell suitable for use is
coronary
artery endothelial cells. Yet other types of vascular endothelial cells
suitable for use

with the present invention include pulmonary artery endothelial cells and
iliac artery
endothelial cells.

In another currently preferred embodiment, suitable endothelial cells can be
obtained from non-vascular tissue. Non-vascular tissue can be derived from any
tubular anatomical structure as described elsewhere herein or can be derived
from
any non-vascular tissue or organ.

In yet another embodiment, endothelial cells can be derived from endothelial
progenitor cells or stem cells; in still another embodiment, endothelial cells
can be
derived from progenitor cells or stem cells generally. In a preferred
embodiment,
the cells can be progenitor cells or stem cells. In other preferred
embodiments, cells

can be non-endothelial cells that are syngeneic, allogeneic, xenogeneic or
autologous derived from vascular or non-vascular tissue or organ. The present
invention also contemplates any of the foregoing which are genetically
altered,
modified or engineered.

In a further embodiment, two or more types of cells are co-cultured to

prepare the present implantable material. For example, a first cell can be
introduced
into the biocompatible matrix and cultured until confluent. The first cell
type can
include, for example, smooth muscle cells, fibroblasts, stem cells,
endothelial
progenitor cells, a combination of smooth muscle cells and fibroblasts, any
other
desired cell type or a combination of desired cell types suitable to create an

21


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
environment conducive to endothelial cell growth. Once the first cell type has
reached confluence, a second cell type is seeded on top of the first confluent
cell
type in, on or within the biocompatible matrix and cultured until both the
first cell
type and second cell type have reached confluence. The second cell type may

include, for example, endothelial cells or any otlzer desired cell type or
combination
of cell types. It is contemplated that the first and second cell types can be
introduced
step wise, or as a single mixture. It is also contemplated that cell density
can be
modified to alter the ratio of smootli muscle cells to endothelial cells.
Similarly,
matrices can be seeded initially with a mixture of different cells suitable
for the

intended indication or clinical regimen.

All that is required of the anchored and/or embedded cells of the present
invention is that they exhibit one or more preferred phenotypes or functional
properties. The present invention is based on the discovery that a cell having
a
readily identifiable phenotype (described elsewhere herein) w11en associated
with a

preferred matrix can reduce, ameliorate, and/or otherwise modulate an immune
response or inflammatory reaction via systemic and/or local effects.

For purposes of the present invention, one such preferred, readily
identifiable
phenotype typical of cells of the present invention is an altered immunogenic
phenotype as measured by the in vitro assays described elsewhere herein.
Another

readily identifiable phenotype typical of cells of the present invention is an
ability to
block or interfere with dendritic cell maturation as measured by the in vitro
assays
described elsewhere herein. Each phenotype is referred to herein as an
immunomodulatory phenotype.

22


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Evaluation of Immunomodulatory Functionality: For purposes of the
invention described herein, the implantable material can be tested for indicia
of
immunomodulatory functionality prior to implantation. For example, samples of
the
implantable material are evaluated to ascertain their ability to reduce
expression of

MHC class II molecules, to reduce expression of co-stimulatory molecules, to
inhibit
the maturation of co-cultured dendritic cells, and to reduce the proliferation
of T
cells. In certain preferred embodiments, the implantable material can be used
for the
purposes described herein when the material is able to reduce expression of
MHC
class II molecules by at least about 25-80%, preferably 50-80%, most
preferably at

least about 80%; to reduce expression of co-stimulatory molecules by at least
about
25-80%, preferably 50-80%, most preferably at least about 80%; inhibit
maturation
of co-cultured dendritic cells by at least about 25-95%, preferably 50-95%,
most
preferably at least about 95%; and/or reduce proliferation of lymphocytes by
at least
about 25-90%, preferably 50-90%, most preferably at least about 90%.

Levels of expression of MHC class II molecules and co-stimulatory
molecules can be quantitated using routine flow cytometry analysis, described
in
detail below. Proliferation of lyinphocytes can be quantitated by in-vitro
coculturing
3[H]-thymidine-labeled CD3+-lymphocytes with the implantable composition via
scintillation-counting as described below in detail. Inhibition of dendritic
cell

maturation can be quantitated by either co-culturing the implantable material
with
dendritic cells and evaluating surface expression of various markers on the
dendritic
cells by flow cytometry and FACS analysis, or by measuring dendritic cell
uptake of
FITC-conjugated dextran by flow cytometry. Each of these methods is described
in
detail below.

23


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
In a typical operative embodiment of the present invention, cells need not
exhibit more than one of the foregoing phenotypes. In certain embodiments,
cells

can exhibit more than one of the foregoing phenotypes.

While the foregoing phenotypes each typify a functional endothelial cell,
such as but not limited to a vascular endothelial cell, a non-endothelial cell
exhibiting such a phenotype(s) is considered endothelial-like for purposes of
the
present invention and thus suitable for use with the present invention. Cells
that are
endothelial-like are also referred to herein as functional analogs of
endothelial cells;
or functional mimics of endothelial cells. Thus, by way of example only, cells

suitable for use with the materials and methods disclosed herein also include
stem
cells or progenitor cells that give rise to endothelial-like cells; cells that
are non-
endothelial cells in origin yet perform functionally like an endothelial cell
using the
parameters set forth herein; cells of any origin which are engineered or
otherwise
modified to have endothelial-like functionality using the parameters set forth
herein.

Typically, cells of the present invention exhibit one or more of the
aforementioned phenotypes when present in confluent, near-confluent or post-
confluent populations and associated with a preferred biocompatible matrix
such as
those described elsewhere herein. As will be appreciated by one of ordinary
skill in
the art, confluent, near-confluent or post-confluent populations of cells are

identifiable readily by a variety of techniques, the most common and widely-
accepted of which is direct microscopic examination. Otliers include
evaluation of
cell number per surface area using standard cell counting techniques such as
but not
limited to a hemocytometer or coulter counter.

24


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Additionally, for purposes of the present invention, endothelial-like cells
include but are not limited to cells which emulate or mimic fiuictionally and
phenotypically confluent, near-confluent or post-confluent endothelial cells
as
measured by the parameters set forth herein.

Thus, using the detailed description and guidance set forth below, the
practitioner of ordinary skill in the art will appreciate how to make, use,
test and
identify operative embodiments of the implantable material disclosed herein.
That
is, the teachings provided herein disclose all that is necessary to make and
use the
present invention's implantable materials. And further, the teachings provided

herein disclose all that is necessary to identify, make and use operatively
equivalent
cell-containing compositions. At bottom, all that is required is that
equivalent cell-
containing compositions are effective to modulate an immune response in
accordance with the methods disclosed herein. As will be appreciated by the
skilled
practitioner, equivalent embodiments of the present composition can be
identified

using only routine experimentation together with the teachings provided
herein.
In certain preferred embodiments, endothelial cells used in the implantable
material of the present invention are isolated from the aorta of human cadaver
donors. Each lot of cells is derived from a single or multiple donors, tested
extensively for endothelial cell purity, biological function, the presence of
bacteria,

fungi, known human pathogens and other adventitious agents. The cells are
cryopreserved and banked using well-known techniques for later expansion in
culture for subsequent formulation in biocompatible implantable materials. In
other
embodiments, living cells can be harvested from a donor or from the patient
for
whom the implantable material is intended.



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Cell Preparation. As stated above, suitable cells can be obtained from a
variety of tissue types and cell types. In certain preferred embodiments,
human
aortic endothelial cells used in the implantable material are isolated from
the aorta of

cadaver donors. In other embodiments, porcine aortic endothelial cells (Cell

Applications, San Diego, CA) are isolated from normal porcine aorta by a
similar
procedure used to isolate human aortic endothelial cells. Each lot of cells is
derived
from a single or multiple donors, tested extensively for endothelial cell
viability,
purity, biological function, the presence of mycoplasma, bacteria, fungi,
yeast,
known human pathogens and other adventitious agents. The cells are further

expanded, characterized and cryopreserved to form a working cell bank at the
tliird
to sixth passage using well-known techniques for later expansion in culture
and for
subsequent formulation as biocompatible implantable material.

The following is an exemplary protocol for preparing endothelial cells
suitable for use with the present invention. Human or porcine aortic
endothelial

cells are prepared in T-75 flasks pre-treated by the addition of approximately
15 ml
of endothelial cell growth media per flask. Human aortic endothelial cells are
prepared in Endothelial Growth Media (EGM-2, Cambrex Biosciences, East
Rutherford, NJ). EGM-2 consists of Endothelial Cell Basal Media (EBM-2,
Cambrex Biosciences) supplemented with EGM-2 which contain 2% FBS. Porcine

cells are prepared in EBM-2 supplemented with 5% FBS and 50 g/ml gentamicin.
The flasks are placed in an incubator maintained at approximately 37 C and 5%
CO2
/ 95% air, 90% humidity for a minimum of 30 minutes. One or two vials of the
cells
are removed from the -160 C -140 C freezer and thawed at approximately 37 C.
Each vial of thawed cells is seeded into two T-75 flasks at a density of

26


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
approximately 3 x 103 cells per cm3, preferably, but no less than 1.0 x 103
and no
more than 7.0 x 103; and the flasks containing the cells are returned to the
incubator.
After about 8-24 hours, the spent media is removed and replaced with fresh
media.
The media is changed every two to three days, thereafter, until the cells
reach

approximately 85-100% confluence preferably, but no less than 60% and no more
than 100%. When the implantable material is intended for clinical application,
only
antibiotic-free media is used in the post-thaw culture of human aortic
endothelial
cells and manufacture of the implantable material of the present invention.

The endothelial cell growth media is then removed, and the monolayer of
cells is rinsed with 10 ml of HEPES buffered saline (HEPES). The HEPES is
removed, and 2 ml of trypsin is added to detach the cells from the surface of
the T-
75 flask. Once detachment has occurred, 3 ml of trypsin neutralizing solution
(TNS)
is added to stop the enzymatic reaction. An additional 5 ml of HEPES is added,
and
the cells are enumerated using a hemocytometer. The cell suspension is
centrifuged

and adjusted to a density of, in the case of human cells, approximately 1.75 x
106
cells/ml using EGM-2 without antibiotics, or in the case of porcine cells,
approximately 1.50 x 106 cells/ml using EBM-2 supplemented with 5% FBS and 50
g/ml gentamicin.

Biocompatible Matrix. According to the present invention, the implantable
material comprises a biocompatible matrix. The matrix is permissive for cell
growth, and cell anchoring to and/or embedding within the matrix. A
particularly
preferred matrix is one characterized by a three-dimensional configuration
such that
anchored and/or embedded cells can create and occupy a multi-dimensional
habitat.
Porous matrices are preferred. The matrix can be a solid or a non-solid.
Certain

27


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
non-solid matrices are flowable and suitable for administration via injection-
type or
infusion-type methods. In certain embodiments, the matrix is flexible and
confonnable. The matrix also can be in the form of a flexible planar form. The
matrix also can be in the form of a gel, a foam, a suspension, a particle, a

microcarrier, a microcapsule, or a fibrous structure. In certain preferred
embodiments, non-solid forms of matrix to which cells are anchored and/or in
which
cells are embedded can be injected or infused when administered.

One currently preferred matrix is Gelfoam (Pfizer, New York, NY), an
absorbable gelatin sponge (hereinafter "Gelfoam matrix"). Gelfoam matrix is a
porous and flexible sponge-like matrix prepared from a specially treated,
purified
porcine dermal gelatin solution.

According to another embodiment, the biocompatible matrix material can be
a modified matrix material. Modifications to the matrix material can be
selected to
optimize and/or to control function of the cells, including the cells'
phenotype (e.g.,

the immunomodulatory phenotype) as described elsewhere herein, when the cells
are
associated with the matrix. According to one embodiment, modifications to the
matrix material include coating the matrix with attachment factors or adhesion
peptides. Exemplary attachment factors include, for example, fibronectin,
fibrin gel,

and covalently attached cell adhesion ligands (including for example RGD)
utilizing
standard aqueous carbodiimide chemistry. Additional cell adhesion ligands
include
peptides having cell adhesion recognition sequences, including but not limited
to:
RGDY, REDVY, GRGDF, GPDSGR, GRGDY and REDV.

28


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
According to another embodiment, the matrix is a matrix other than

Gelfoam. Additional exemplary matrix materials include, for example, fibrin
gel,
alginate, polystyrene sodium sulfonate microcarriers, collagen coated dextran
microcarriers, cellulose, PLA/PGA and pHEMA/MMA copolymers (with polymer

ratios ranging from 1-100% for each copolymer). According to a preferred
embodiment, these additional matrices are modified to include attachment
factors, as
recited and described above.

According to another embodiment, the biocompatible matrix material is
physically modified to improve cell attachment to the matrix. According to one
embodiment, the matrix is cross linked to enhance its mechanical properties
and to

improve its cell attachment and growth properties. According to a preferred
embodiment, an alginate matrix is first cross linked using calcium sulfate
followed
by a second cross linlcing step using calcium chloride and routine protocols.

According to yet another embodiment, the pore size of the biocompatible
matrix is modified. A currently preferred matrix pore size is about 25 m to
about
100 m; preferably about 25 m to 50 m; more preferably about 50 m to 75 m;
even more preferably about 75 m to 100 m. Other preferred pore sizes include
pore sizes below about 25 m and above abolut 100 m. According to one

embodiment, the pore size is modified using a salt leaching technique. Sodium
cliloride is mixed in a solution of the matrix material and a solvent, the
solution is
poured into a mold, and the solvent is allowed to evaporate. The matrix/salt
block is
then immersed in water and the salt leached out leaving a porous structure.
The
solvent is chosen so that the matrix is in the solution but the salt is not.
One
exemplary solution includes PLA and methylene chloride.

29


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
According to an alternative embodiment, carbon dioxide gas bubbles are
incorporated into a non-solid form of the matrix and then stabilized with an
appropriate surfactant. The gas bubbles are subsequently removed using a
vacuum,
leaving a porous structure.

According to another embodiment, a freeze-drying technique is employed to
control the pore size of the matrix, using the freezing rate of the ice
microparticles to
form pores of different sizes. For example, a gelatin solution of about 0.1-2%

porcine or bovine gelatin can be poured into a mold or dish and pre-frozen at
a
variety of different temperatures and then lyophilized for a period of time.
The

material can then be cross-linked by using, preferably, ultraviolet light (254
nm) or
by adding gluteraldehyde (formaldehyde). Variations in pre-freezing
temperature
(for example -20 C, -80 C or -180 C), lyophilizing temperature (freeze dry at
about
-50 C), and gelatin concentration (0.1% to 2.0%; pore size is generally
inversely
proportional to the concentration of gelatin in the solution) can all affect
the

resulting pore size of the matrix material and can be modified to create a
preferred
material. The skilled artisan will appreciate that a suitable pore size is
that which
promotes and sustains optimal cell populations having the phenotypes described
elsewhere herein.

Cell Seeding of Biocompatible Matrix. The following is a description of one
exemplary configuration of a biocompatible matrix. As stated elsewhere,
preferred
matrices are solid or non-solid, and can be formulated for implantation,
injection or
infusion.



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Pre-cut pieces of a suitable biocompatible matrix or an aliquot of suitable
biocompatible flowable matrix are re-hydrated by the addition of EGM-2 without
antibiotics at approximately 37 C and 5% CO2 / 95% air for 12 to 24 hours. The
implantable material is then removed from their re-hydration containers and
placed

in individual tissue culture dishes. Biocompatible matrix is seeded at a
preferred
density of approximately 1.5-2.0 x 105 cells (1.25-1.66 x 105 cells/cm3 of
matrix)
and placed in an incubator maintained at approximately 37 C and 5% CO2 / 95%
air,
90% humidity for 3-4 hours to facilitate cell attachment. The seeded matrix is
then
placed into individual containers (Evergreen, Los Angeles, CA) tubes, each
fitted

with a cap containing a 0.2 gm filter with EGM-2 and incubated at
approximately
37 C and 5% CO2 / 95% air. The media is changed every two to three days,
thereafter, until the cells have reached confluence. The cells in one
preferred
embodiment are preferably passage 6, but cells of fewer or more passages can
be
used.

Cell Growth. A sample of implantable material is removed on or around
days 3 or 4, 6 or 7, 9 or 10, and 12 or 13, the cells are counted and assessed
for
viability, and a growth curve is constructed and evaluated in order to assess
the
growth characteristics and to determine whether confluence, near-confluence or
post-confluence has been acliieved. Generally, one of ordinary skill will
appreciate

the indicia of acceptable cell growth at early, mid- and late time points,
such as
observation of an exponential increase in cell number at early time points
(for
example, between about days 2-6 when using porcine aortic endothelial cells),
followed by a near confluent phase (for example, between about days 6-8),
followed

by a plateau in cell number once the cells have reached confluence (for
example,
31


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
between about days 8-10) and maintenance of the cell number when the cells are
post-confluent (for example, between about days 10-14).

Cell counts are achieved by complete digestion of the aliquot of implantable
material with a solution of 0.5 mg/ml collagenase in a HEPES/Ca}+ solution.
After
measuring the volume of the digested implantable material, a known voluine of
the
cell suspension is diluted with 0.4% trypan blue (4:1 cells to trypan blue)
and

viability assessed by trypan blue exclusion. Viable, non-viable and total
cells are
enumerated using a hemocytometer. Growth curves are constructed by plotting
the
number of viable cells versus the number of days in culture.

For purposes of the present invention, confluence is defined as the presence
of at least about 4 x 105 cells/cm3 when in an exemplary flexible planar form
of the
implantable material (1.0 x 4.0 x 0.3 cm), and preferably about 7 x 105 to 1 x
106
total cells per aliquot (50-70 mg) when in an injectable or infusable
composition.
For both, cell viability is at least about 90% preferably but no less than
80%.

An exemplary embodiment of the present invention comprises a
biocompatible matrix and cells suitable for use witli any one of the various
clinical
indications or treatment paradigms described herein. Specifically, in one
preferred
embodiment, the implantable material comprises a biocompatible matrix and

endothelial cells, endothelial-like cells, or analogs of either of the
foregoing. In one
currently preferred embodiment, the implantable material is in a flexible
planar form
and comprises endothelial cells, preferably vascular endothelial cells such as
but not
limited to human aortic endotllelial cells and the biocompatible matrix
Gelfoam
gelatin sponge (Pfizer, New York, NY, hereinafter "Gelfoam matrix").

32


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Implantable material of the present invention comprises cells anchored to
and/or embedded within a biocompatible matrix. Anchored to and/or embedded
within means securedly attached via cell to cell and/or cell to matrix
interactions
such that the cells withstand the rigors of the preparatory manipulations
disclosed

herein. As explained elsewhere herein, an operative embodiment of implantable
material comprises a near-confluent, confluent or post-confluent cell
population
having a preferred phenotype. It is understood that embodiments of implantable
material likely shed cells during preparatory manipulations and/or that
certain cells
are not as securedly attached as are other cells. All that is required is that

implantable material comprise cells that meet the functional or phenotypical
criteria
set forth elsewhere herein.

The implantable material of the present invention was developed on the
principals of tissue engineering and represents a novel approach to addressing
the
herein-described clinical needs. The implantable material of the present
invention is

unique in that the viable cells anchored to and/or embedded within the
biocompatible matrix are able to supply to the site of administration multiple
cell-
based products in physiological proportions under physiological feed-back
control.
As described elsewhere herein, the cells suitable for use with the implantable

material are endothelial, endothelial-like cells, or analogs of each of the
foregoing.
Local delivery of multiple compounds by these cells and physiologically-
dynamic
dosing provide more effective regulation of the processes responsible for
modulating
an immune response. The implantable material of the present invention can
provide
an environment which mimics supportive physiology and is conducive to
modulation of an immune response.

33


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Evaluation of Functionality. For purposes of the invention described herein,
the implantable material is tested for indicia of functionality prior to
delivery to a
recipient. For example, as one determination of suitability, conditioned media
are
collected during the culture period to ascertain levels of heparan sulfate or

transforming growth factor-(31 (TGF-(31) or basic fibroblast growth factor (b-
FGF)
or nitric oxide which are produced by the cultured endothelial cells. In
certain
preferred embodiments, the implantable material can be used for the purposes
described herein when total cell number is at least about 1, preferably about
2, more

preferably at least about 4 x 105 cells/cm3 of flexible planar form;
percentage of

io viable cells is at least about 80-90%, preferably >90%, most preferably at
least about
90%; heparan sulfate in conditioned media is at least about 0.1-0.5 preferably
at
least about 0.23 microg/mL/day. If other indicia are desired,,then TGF-(31 in
conditioned media is at least about 200-300, preferably at least about 300
picog/ml/day; b-FGF in conditioned media is below about 200 picog/ml,
preferably
no more than about 400 picog/ml.

Heparan sulfate levels can be quantitated using a routine dimethylmethylene
blue-chondroitinase ABC digestion spectrophotometric assay. Total sulfated
glycosaminoglycan (GAG) levels are determined using a dimethylmethylene blue
(DMB) dye binding assay in which unknown samples are compared to a standard

curve generated using known quantities of purified chondroitin sulfate diluted
in
collection media. Additional samples of conditioned medium are mixed with
chondroitinase ABC to digest chondroitin and dermatan sulfates prior to the
addition
of the DMB color reagent. All absorbances are determined at the maximum
wavelength absorbance of the DMB dye mixed with the GAG standard, generally

34


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
around 515-525 nm. The concentration of heparan sulfate per day is calculated
by
subtracting the concentration of chondroitin and dermatan sulfate from the
total
sulfated glycosaminoglycan concentration in conditioned medium samples.
Chondroitinase ABC activity is confirmed by digesting a sample of purified

chondroitin sulfate. Conditioned medium samples are corrected appropriately if
less
than 100% of the purified chondroitin sulfate is digested. Heparan sulfate
levels
may also be quantitated using an ELISA assay employing monoclonal antibodies.

If desired, TGF-(31 and b-FGF levels can be quantitated using an ELISA
assay employing monoclonal or polyclonal antibodies, preferably polyclonal.
lo Control collection media can also be quantitated using an ELISA assay and
the

samples corrected appropriately for TGF-(31 and b-FGF levels present in
control
media. Nitric oxide (NO) levels can be quantitated using a standard Griess
Reaction
assay. The transient and volatile nature of nitric oxide makes it unsuitable
for most
detection methods. However, two stable breakdown products of nitric oxide,
nitrate

(NO3) and nitrite (NOZ), can be detected using routine photometric methods.
The
Griess Reaction assay enzymatically converts nitrate to nitrite in the
presence of
nitrate reductase. Nitrite is detected colorimetrically as a colored azo dye
product,
absorbing visible light in the range of about 540 nm. The level of nitric
oxide
present in the system is determined by converting all nitrate into nitrite,
determining

the total concentration of nitrite in the unknown samples, and then comparing
the
resulting concentration of nitrite to a standard curve generated using known
quantities of nitrate converted to nitrite.



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Also, any one or more of the foregoing assays can be used alone or in
combination as a screening assay for identifying a cell as suitable for use
with the
implantable material of the present invention.

While the earlier-described preferred immunomodulatory phenotype can be
assessed using one or more of the optional quantitative heparin sulfate, TGF-
(31, NO
and/or b-FGF functional assays described above, implantable material can be
evaluated for the presence of one or more preferred immunomodulatory
phenotypes
as follows. For purposes of the present invention, one such preferred, readily
identifiable phenotype typical of cells of the preseiit invention is an
altered

immunogenic phenotype as measured by the in vitro assays described below.
Another readily identifiable phenotype typical of cells of the present
invention is an
ability to block or interfere with dendritic cell maturation as measured by
the in vitro
assays described below. Each plienotype is referred to herein as an

immunomodulatory phenotype and cells exhibiting such a phenotype have
immunomodulatory functionality.

Evaluation of Immunomodulatory Functionality: For purposes of the
invention described herein, the immunomodulatory functionality of implantable
material can be tested as follows. For example, samples of the implantable
material
are evaluated to ascertain their ability to reduce expression of MHC class II

molecules, to reduce expression of co-stimulatory molecules, to inhibit the
maturation of co-cultured dendritic cells, and to reduce the proliferation of
T cells.
In certain preferred embodiments, the implantable material can be used for the
purposes described herein when the material is able to reduce expression of
MHC
class II molecules by at least about 25-80%, preferably 50-80%, most
preferably at

36


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
least about 80%; to reduce expression of co-stimulatory molecules by at least
about
25-80%, preferably 50-80%, most preferably at least about 80%; inhibit
maturation
of co-cultured dendritic cells by at least about 25-95%, preferably 50-95%,
most
preferably at least about 95%; and/or reduce proliferation of lymphocytes by
at least

about 25-90%, preferably 50-90%, most preferably at least about 90%.
Levels of expression of MHC class II molecules and co-stimulatory
molecules can be quantitated using routine flow cytometry and FACS-analysis,
described in detail below. Proliferation of lymphocytes can be quantitated can
be
quantitated by in-vitro coculturing 3[H]-thymidine-labeled CD3+-lymphocytes
with

the implantable composition via scintillation-counting as described below in
detail.
Iiihibition of dendritic cell maturation can be quantitated by either co-
culturing the
implantable material with dendritic cells and evaluating surface expression of
various markers on the dendritic cells by flow cytometry and FACS analysis, or
by
measuring dendritic cell uptake of FITC-conjugated dextran by flow cytometry.

Each of these methods is described in detail below.

Also, any one or more of the foregoing assays can be used alone or in
combination as a screening assay for identifying a cell as suitable for use
with the
implantable material of the present invention.

Methods of Use and Clinical Indications: This invention is directed

generally to materials and methods for modulating an immunologically adverse
response, including an inflammatory reaction, to an exogenous immunogen or
stimulus as well as an endogenous iinmunogen or stimulus. The invention is
also
directed to a cell-, tissue-, or organ-associated immunogen. For example, the
present invention can modulate an adverse immune response to non-syngeneic or

37


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
syngeneic cells, tissues or organs and/or ameliorate a pre-existing immune
condition
such as but not limited to an autoimmune condition. This discussion of
implantable
materials and methods of use for suitable clinical indications will make
reference to
the following terms and concepts.

An early phase immune response depends on innate immunity. During an
innate immune response, a variety of innate immune mechanisms recognize and
respond to the presence of immunogen. Innate immunity is present in all
individuals
at all times and principally discriminates between self, altered self and non-
self. For
example, a type of innate immune cell is the Natural Killer (NK) cell, the
dendritic

cell and the monocyte. The innate immune response is followed by an adaptive
immune response, mediated by clonal selection of specific lymphocytes and
resulting in a more tailored and long-lasting immune response against the
recognized
antigen.

The adaptive immune response, or adaptive iminunity, is the response of
i5 antigen-specific lymphocytes to antigen, including the development of
immunological memory. Adaptive immune responses are generated by clonal
selection of lymphocytes. Adaptive immune responses are distinct from innate
and
non-adaptive phases of immunity, which are not mediated by clonal selection of
antigen-specific lympliocytes. The adaptive immune response includes both cell-


mediated immunity and huinoral immunity. For example, an adaptive immune cell
is a B-cell or T-cell lymphocyte.

One of the hallmarks of an adaptive immune response is establishment of
immunological memory. Immunological memory is the ability of the immune
system to respond more rapidly and effectively to immunogens been encountered

38


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
previously, and reflects the pre-existence of a clonally expanded population
of
antigen-specific lymphocytes.

Protective immunity can be either cell-mediated immunity or humoral
immunity. Humoral immunity is specific immunity mediated by antibodies made in
a humoral immune response. Cell-mediated immunity describes any adaptive

immune response in which antigen specific T cells play a main role.

Autoimmune diseases are mediated by sustained adaptive immune responses
specific for self antigens. Tissue injury results because the antigen is an
intrinsic
component of the body and consequently effector mechanisms of the immune

system are directed at self tissues. Also, since the offending autoantigen can
not be
removed from the body, the immune response persists, and there is a constant
supply
of new autoantigen, which amplifies the response.

Altliough some syngeneic grafts or transplants may be accepted long-term,
even syngeneic grafts can be problematic for a recipient. In fact, even when

autologous cells are harvested, manipulated ex vivo and returned to the
original
donor, non-acceptance may occur to some extent. Typically, grafts differing at
the
MHC or at other genetic loci are rejected in the short term by a recipient T-
cell
response. When donor and recipient differ at the MHC, for example, the immune
response is directed at the non-self MHC molecule or other surface molecules

expressed by the graft. Acceptance or rejection of a graft or transplant
invokes
immune events such as antigen recognition, T-cell activation, T-helper cell
recruitment and ultimately graft destruction.

39


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
An inflammatory reaction is initiated by a local immune response. Acute
inflammation is an early transient episode, while clironic inflammation
persists such
as during autoimmune responses. Inflammation reflects the effects of cytokines
on
local blood vessels. Cytokines have important effects on the adherent
properties of

the blood vessel endothelium, causing circulating leukocytes to stick to the
endothelial cells of the blood vessel wall and migrate through the wall. Later-
stage
inflammatory responses also involve lymphocytes of the adaptive iminune
response
which have been activated by immunogen.

Exemplary methods of treatment and clinical indications are discussed

below. This is not intended to be an exhaustive discussion. The present
invention
contemplates any clinical indication suitable for treatnient with the present
invention, including any clinical indication typified by or otherwise
associated with
an immunological event having adverse clinical consequences for a patient.

Syngeneic and Non-syngeneic Transplants: The present invention can be
used to reduce or diminish a transplant recipient's adverse response to a
cell, tissue
and/or organ transplant, whether it be a syngeneic or a non-syngeneic
transplant.
The present invention can also be used to stabilize or maintain a transplant
recipient's acceptance of a cell, tissue or organ transplant, whether it be a
syngeneic
or a non-syngeneic transplant. As taught herein, modulation of an adverse
imnlune

response occurs when implantable material is used as a pre-transplant
treatment,
coincident treatment or post-transplant treatment. For example, it is
contemplated
that a pre-treatment can acclimate a recipient's immune system which
facilitates
later acceptance of the transplant. Similarly, coincident treatment can
shorten the
time course of physiological events which ultimately result in acceptance and



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
ameliorate any adverse immunological events provoked by the transplant. Post-
transplant treatments, whether single or multiple, can perpetuate a state of
acceptance and keep adverse immunological events in check if/when such events
occur. Clinically, typical indications suitable for treatment with the
implantable

material of present invention include, but are not limited to, allorejection,
xenorejection, ischemia-reperfusion injury associated with transplanted
tissues or
organs, and repetitive treatment courses. Repetitive treatment courses
include, for
exainple, recurrent atherosclerosis at different vessel sites requiring
repetitive
intervention and repetitive replenishing injections of pancreas islet cells.
For

purposes of the present invention, blood is a type of tissue and blood
transfusion
recipients can benefit from treatment with the present invention for all the
foregoing
reasons. Similarly, immunological-based diseases associated with cell, tissue
and/or
organ transplants benefit from the treatment paradigms set forth above.

Complement dependent Cytotoxicity: In addition to reducing, modulating or
eliminating the innate immune response and/or the adaptive immune response, as
outlined above, the implantable material of the present invention can also
reduce,
modulate or eliminate the severity of the complement cascade and the
inflammatory
side effects of complement activation. For example, attenuation of the
complement
cascade using the implantable material or the present invention reduces
complement

mediated cell lysis of a transplanted tissue or organ, thereby ameliorating
transplant
dysfunction and extending the duration of successful treatment.

Interventional Therapies: As taught herein, the present invention can
modulate the severity or robustness of an already-existing immune response as
well
as a future response provoked by subsequent earlier exposure(s) to an
immunogen.
41


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Under such circumstances, implantable material can intervene by blocking

escalation of an adverse immune response or diminishing onset of
hypersensitivity,
respectively. Suppression of a memory response can avoid further physiological
insult which can jeopardize a patient's organ health, for example. In the case
of an

already-existing condition, such as an auto-immune condition, the present
inverition
can quell the devastating effects of unabated immunological assaults on a
patient's
tissues or organs. In essence, such patients are continuously exposed to
offending
immunogen and their immune response escalates out-of-control resulting in
serious,
often fatal, disease sequelae.

While an auto-immune condition can be likened to serial challenges with an
offending immunogen, other clinical indications can be considered similarly.
For
example, as suggested above, a recipient of a syngeneic or non-syngeneic
transplant
is subject to serial challenges. Replenishment of a transplant, such as kidney
islet
cells which deteriorate over time, constitutes a serial challenge. Secondary

infarctions or secondary vascular injuries can be considered serial
challenges.
Another example is a disease such as but not limited to vasculitis. Any of the
foregoing can be effectively managed using the materials and methods of the
present
invention.

Supplanting Immunosuppressive Agents: As explained elsewhere herein, it
is contemplated that administration of the implantable material of the present
invention inhibits sufficiently at least T cell activation such that the need
to
administer haxmful immunosuppressive agents is eliminated or significantly
reduced. However, it is also contemplated that a certain class of patients,
such as a
patient pre-disposed to highly exacerbated immune responses, can be treated
with

42


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
both implantable material and an immunosuppressive agent. The implantable
material of the present invention, when administered prior to or coincident
with
transplantation of syngeneic or non-syngeneic tissue, can permit reduced
dosages of
immunosuppressive agent, if one is necessary, to manage a potential graft
rejection
response.

Potent immunosuppressive agents, for example, cyclosporin A, tacrolimus
(FK-506), sirolimus (rapamicin), mycophenolate mofetil, leflunomide,
glucocorticoids, cytostatics, azathioprine, and prednisone, are administered
to a
transplant recipient to inhibit T cell activation and increase the probability
of graft

survival. However, administration of potent immunosuppressive agents increases
the risk of cancer and infection and contributes to the risk of other side
effects
including hypertension, dyslipidemia, hyperglycemia, peptic ulcers, and liver
and
kidney injuries. The present invention can permit more prudent and less risky
dosing regimens of such agents. Additionally, immunosuppressants which are

typically administered to an organ recipient can be administered prior to,
coincident
with and/or subsequent to administration of the implantable material of the
present
invention. For example, implantable materials can amplify the beneficial
effects of
immunosuppressants while minimizing the risks of such agents in recipients
whose
immune system is overstimulated or over-sensitized, perhaps reducing the time
in

which immunomodulation is actually achieved. It is further contemplated that
dosages of immunosuppressants, in certain embodiments, are less than those
typically administered in the absence of implantable material, thereby
exposing a
recipient to less toxic doses of immunosuppressants.

43


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Altering the Time Course of an Immune Response: In a preferred
embodiment of the invention, matrix-anchored and/or embedded endothelial cells

are administered to diminish or delay an immune or inflammatory response. It
is not
necessary that the implantable material completely eliminate an immune or

inflammatory response to be considered effective. Rather, the material need
only
alter the time course of a response, such as by reducing the duration of an
immune or
inflainmatory response or by reducing an acute inflammatory response to a
chronic
inflammatory response. Delaying an immune or inflammatory response allows a
coincident or later administered therapy to effectively treat a recipient in
the absence

of an immune or inflammatory response and/or to increase the duration of
transplant
acceptance. Thus any delay or reduction in the severity of an adverse immune
response is beneficial clinically to a patient.

Furtliermore, the implantable material of the present invention can also be
used to manage or reduce an immune response and inflammatory reaction
associated
with any exogenous foreign body or foreign material introduced to a patient,
or any
form of exogenous stimulus. The present invention contemplates exogenous

immunogens which are naturally-occurring. The present invention also
contemplates exogenous immunogens, including but not limited to pharmaceutical
agents, toxins, surgical implants, infectious agents and chemicals. For
purposes of

the present invention, an exogenous immunogen can be an exogenous stimulus
such
as, but not limited to, environmental stress, injury, exposure or any stimulus
which
provokes an adverse immune response or inflammatory reaction.

44


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
For example, synthetic graft materials, such as a synthetic PTFE
arteriovenous graft, or other synthetic surgical materials or prosthetic
devices, can
induce a foreign body reaction in the host. This type of immune or
inflammatory
response can also be reduced or eliminated by administering the implantable

material of the present invention to the patient prior to or at the time of
implanting
the synthetic material. Administration subsequent to implantation is also
effective.
Reducing any foreign body reaction in the host improves the overall function
and/or
outcome of the treatment.

General Considerations. In certain embodiments of the invention, additional
therapeutic agents are administered prior to, coincident with and/or following
administration of the implantable material. For example, cytokines or growth
factors can also be incorporated into the implantable material, depending on
the
clinical indication necessitating the implant, including agents which can mute
an
immune-related humoral or cellular event, or tissue-associated biochemical
cascade.

Other types of therapeutic agents include those which can promote the
longevity of
cells anchored to and/or embedded within the implantable material and/or
agents
which can delay the bioerosion of an erodible biocompatible matrix post
implantation. Any of the foregoing can be administered locally or
systemically; if
locally, certain agents can be contained within the implantable material or

contributed by the cells per se.

Administration Considerations. As contemplated herein, the implantable
material of the present invention can be delivered to or situated at any
compatible
anatomical site provided that conditions at the site do not cause mechanical-
type or
physical-type disruption or untimely disintegration of the implantable
material, or


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
otherwise compromise the physical integrity or the ftmctionality of the
iinplantable
material. For example, the present invention can be situated subcutaneously,
perivascularly, or intraperitoneally. One preferred site is a skin pouch.
Other
preferred sites can be perivascular or non-perivascular. The implantable
material can

be situated adjacent to or in contact with an organ or a tubular anatomical
structure
which can be a vascular or non-vascular structure. The present invention can
be
delivered to any coinpatible site for purposes of either systemic modulation
of a
humoral or cellular immune response, or for purposes of local modulation of an
inflammatory reaction, or both. Certain preferred embodiments of implantable

io material can reside at an implantation site for at least about 56-84 days,
preferably
about at least 7 days, more preferably about at least 14 days, even more
preferably
about at least 28 days, and most preferably more than about 28 days before it
bioerodes.

When ready for delivery to a recipient, the implantable material when in an
exemplary flexible planar form, is a 1 x 4 x 0.3 cm (1.2 cm3) sterile piece
with
preferably approximately 5-8 x 105 preferably at least about 4 x 105 cells/cm3
and at
least about 90% viable cells, for example, human aortic endothelial cells
derived
from a single cadaver donor source, per cubic centimeter in approximately 45-
60 ml,
preferably about 50 ml, endothelial growth medium (for example, endothelial

growth medium (EGM-2) containing no phenol red and no antibiotics. When
porcine aortic endothelial cells are used, the growth medium is also EBM-2
containing no phenol red, but supplemented with 5% FBS and 50 g/ml
gentamicin.

46


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
In certain embodiments contemplated herein, the iinplantable material of the
present invention is a flowable composition comprising a particulate
biocompatible
matrix which can be in the form of a gel, a foam, a suspension, a particle, a

microcarrier, a microcapsule, or other flowable material. Any non-solid
flowable
composition for use with an injection-type or infusion-type delivery device is
contemplated herein. In certain embodiments, the flowable composition is
preferably a shape-retaining composition. An implantable material comprising
cells
in, on or within a flowable-type particulate matrix as contemplated herein can
be
formulated for use with any injection-type delivery device ranging in internal

diameter from about 22 gauge to about 26 gauge and capable of delivering about
50
mg of flowable composition comprising particulate material containing
preferably
about 1 million cells in about 1 to about 3 ml.

According to a currently preferred embodiment, the flowable composition
comprises a biocompatible particulate matrix such as Gelfoam particles,
Gelfoam
powder, or pulverized Gelfoam (Pfizer Inc., New York, NY) (llereinafter
"Gelfoam

particles"), a product derived from porcine dermal gelatin. According to
another
embodiment, the particulate matrix is Cytodex-3 (Amersham Biosciences,
Piscataway, NJ) microcarriers, comprised of denatured collagen coupled to a
matrix
of cross-linked dextran.

Endovascular Administration. The flowable composition can also be
administered via an intraluminal or endovascular route even though the final
deposition site is not intraluminal. For example, the composition can be
delivered

by any device able to be inserted within the blood vessel. Endoscopic guidance
systems may be used to locate the delivery device at the site of
administration,
47


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
including, for example, intravascular ultrasound (IVUS), color Doppler
ultrasound,
duplex ultrasound, other routine ultrasound, angiography, magnetic resonance
angiography (1VIRA), magnetic resonance imaging (MRI), CT scanning,
fluoroscopy
to identify the location of a stent and/or other endoscopic guidance systems
known

in the field. Additionally, the site of administration may be located using
tactile
palpation.

In one instance, the intraluminal delivery device is equipped with a
traversing or penetrating device which penetrates the luminal wall of a blood
vessel
to reach a non-luminal surface of a blood vessel. The flowable composition is
then

io deposited on the non-luminal surface. It is contemplated herein that a non-
luminal,
also termed an extraluminal, surface can include any site exterior to a blood
vessel
or any perivascular surface of a vessel, or can be within the adventitia,
media, or
intima of a blood vessel, for example. For purposes of this invention, non-
luminal
or extraluminal means any surface except an interior surface of the lumen. It
is also

contemplated that deposition within the perivascular space can be
accoinplished via
an intraluminal delivery device and does not require contact with the
extraluminal
surface of the traversed vessel.

The penetrating devices contemplated herein can permit, for example, a
single point of delivery or a plurality of delivery points arranged in a
desired

geometric configuration to accomplish delivery of the flowable composition to
a
non-luminal surface of a blood vessel without disrupting an injured or
diseased
target site. A plurality of delivery points can be arranged, for example, in a
circle, a
bulls-eye, or a linear array arrangement to name but a few. The penetrating
device

48


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
can also be in the form of a stent perforator, such as but not limited to, a
balloon
stent including a plurality of delivery points.

Percutaneous Administration. Flowable composition can be delivered via a
percutaneous route using a needle, catheter or other suitable delivery device.
The
flowable composition can be delivered percutaneously coincident with use of a

guidance method to facilitate delivery to the site in need of treatment. The
guidance
step is optional. Endoscopic guidance systems can be used to locate a site of
extraluminal adininistration, for example, including intravascular ultrasound
(IVUS), color Doppler ultrasound, duplex ultrasound, other routine ultrasound,

angiography, magnetic resonance angiography (MRA), magnetic resonance imaging
(MRI), CT scanning fluoroscopy. Additionally, the site of administration can
be
located using tactile palpation. Upon entry into the perivascular or
peritoneal space,
for example, the clinician can deposit the flowable composition on any non-
luminal
surface or at any non-luminal site. The guiding or identifying step is
optionally

performed and not required to practice the methods of the present invention.
In
another embodiment, the implantable flowable composition is delivered locally
to a
surgically-exposed extraluminal site.

Also contemplated is administration by infusion. Infusion can be
accomplished as a bolus-type dose or a slower-type, gradual dose. The skilled

clinician will recognize the advantages of each and will recognize the
circumstances
in wliich to employ one or the other modes of administration. All that is
required is
routine clinical infusion apparatus.

49


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Experimental Materials and Procedures

Material Preparation and Evaluation. As described in greater detail
elsewhere herein, porcine aortic endothelial cells and huinan aortic
endothelial cells
were individually isolated and cultured. The cultured cells were then seeded
on a

three-dimensional biocoinpatible matrix, such as Gelfoam, and incubated until
the
cells reached confluence. The functionality of the endothelial cells anchored
to
and/or embedded within the matrix was evaluated according to the previously
discussed protocols.

Endothelial cell-induced immune reaction in rats. Fifty-four Sprague-Dawley
rats received 5x105 porcine aortic endothelial cell transplants in the
subcutaneous
'dorsal space as Gelfoam-embedded cells, saline-suspended cell pellets, or as
pellets
adjacent to empty Gelfoam. After dorsal incision, a small subcutaneous cavity
was
built in blunt technique and Gelfoam-embedded cells carefully inserted or
cells
injected. Empty control Gelfoam matrices were incubated in complete DMEM prior

to implantation. Sera were collected serially from 0 to 56 days, aliquoted and
stored
at -70 C.

Endothelial cell-induced immune reaction in mice. Thirty-six B6-mice
received 5x105 porcine aortic endothelial cell implants in the subcutaneous
dorsal
space as Gelfoam-embedded cells, saline-suspended cell pellets, or as pellets

2o adjacent to empty Gelfoam. Empty control Gelfoam matrices were incubated in
complete DMEM prior to implantation. To evaluate the impact of matrix-
embedding on immunological memory the same groups of mice were rechallenged
witli the identical treatment on day 100. Sera were collected serially from 0
to 90
days after each implantation-procedure, aliquoted and stored at -70 C. Four
mice of



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
each group were sacrificed on day 28 and day 128 respectively for splenocyte
isolation.

Endothelial cell-induced immune reaction in serially challenged mice.
Porcine aortic endothelial cells (PAE) isolated from LargeWhite swine aorta
were
either seeded on Gelfoain as previously described or grown to confluence on

polystyrene plates. B6-mice received injections in the subcutaneous dorsal
space on
days 0, 21, 35 of 5x105 PAE (n=24, pre-sensitized mice) or saline (n=24, naive
mice). On day 42, 12 mice from each group received 5x105 matrix-embedded or
free PAE. Host immune reactions and lytic dainage of endotlielial cells were

studied for the following 90 days. Sera were collected serially from days 42
to 132,
aliquoted and stored at -70 C. Six mice of each group were sacrificed on day
70,
the remaining on day 132 for splenocyte isolation.

EXPERIMENTS
Endothelial cells embedded in a three-dimensional matrix grow in a three-
dimensional pattern.

Scanning electron microscopy was performed to evaluate the growing pattern
of endothelial cells grown within a biocompatible matrix. Implantable material
comprising endothelial cell anchored to and/or embedded within a Gelfoam
matrix

were rinsed with PBS, divided into 0.5 cm specimens, fixed with 3%
glutaraldehyde
(Sigma Chemicals; St. Louis, MO) 90 min, and transferred to distilled water.
After
incubation in 1 / OsO4, specimens were rinsed with distilled water and
dehydrated
in serial solutions of ethanol (30, 50, 75, 80, 85, 90, 95, and 100%) at 15
min

intervals, and hexamethyldisilazane (Sigma) (50%, 100%) at 30 min intervals.

Specimens were evaporated overnight in 100% HMDS and thereafter coated with
51


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
gold in a plasma coater (Edwards Coating System, U.K.). Scanning electron
microphotographs were obtained at 5-kV acceleration voltage (Stereoscan 240,
Cambridge Instruments, U.K.).

Scanning electron microscopy revealed a 3-D growing pattern of porcine

aortic endothelial cells along the interstices of the Gelfoam-matrix. Cell
viability
remained at 95% over the 2-week culture course.

Experimental data indicate that the in-vivo immunoacceptance of Gelfoam-
embedded cells is an effect of the three-dimensional growing pattern of
endothelial
cells in the matrix rather than from the presence of the biocompatible matrix
alone.
Typically, implanted cells or proteins combined within tissue-engineered

biomaterials serve as a source of antigens immuno-stimulating. Yet, the
Gelfoain
matrix is immunoneutral and itself has no immune protective effect since
injection
of porcine aortic endothelial cells adjacent to Gelfoam matrix alone evoked
the same
immune response as free injected endothelial cells. The nature of endotlielial
cells

contributes to this unique form of immunomodulation observed with matrix-
embedded cell preparations. In particular, these cells have a sidedness: a
basal
surface that interacts with basement membrane and superior surface that
interacts
with flowing blood and cellular elements. Data suggest that endothelial cell
fiuiction is anchorage- and density-dependent. Systemic diseases like
hypertension,

alterations in lipid and glucose metabolism or exposure to toxins alter
anchorage-
dependent regulation and the amplitude and nature of immune responses against
the
endothelium and phenotypic transformation of intact endothelial cells from
matrix-
adherent to free contributes to initiation of vascular disease.

52


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Modulation of surface molecules including co-stimulatory and adhesion
molecules.
Expression levels of costimulatory and adhesion molecules on endothelial

cells in vitro were quantified by flow cytometry. FITC- and PE-labeled
antibodies
were used and included mouse anti-porcine P-selectin antibody, mouse anti-
porcine
CD31 (clone LCI-4), anti-human CD54 (clone 15.2), anti-human CD62E (clone

1.2B6), anti-human CD58 (clone 1C3), anti-human CD80 (clone BB1), anti-human
CD86 (clone 2331), anti-human 4-1BB-ligand (PE-labeled, clone C65-485), rat
anti-
mouse IgGI (clone A85-1), and anti-mouse IgM (clone R6-60.2), rabbit anti-rat
IgG,
rabbit anti-human CD40, goat anti-rabbit IgG, mouse anti-human CD 106 (clone

1.G11B1), mouse anti-human HLA-DP,DQ,DR (clone CR3/43), mouse anti class I
MHC (IgG2a), rat anti-mouse IgG2a, mouse anti-human ox40-ligand, mouse anti-
human Programmed Death Ligand 1(PD-L1, clone MIHl), anti-human PD-L2
(clone MIH18), and anti-human inducible costimulator ligand (ICOS-ligand,
clone
MIH12).

Endothelial cell monolayers or endothelial cells embedded in Gelfoam were
harvested after culture in complete medium (CD3 1, CD58, PD-L2, ox40-ligand,
MHC-I), stimulated witli 100 U/ml TNF-a (CD54, CD80, CD86, CD106, E-
selectin, P-selectin) or 200 U/ml TNF-a (ICOS-L) for 24 hours, 10 g/ml LPS
for
24 hours (4-1BB-ligand), 1000 U/ml IFN-y (MHC-II, CD40), or 100 U/ml IFN-y

and 25 ng/m1 TNF-a (PD-L1) for 48 hours. Media were aspirated and cells were
washed with PBS. Monolayers incubated in 1.0 mM PBS/EDTA for 5 min, and
disrupted by gentle shaking. Gelfoam were digested with collagenase type I,
shown
to have no effect on expression of surface molecules. Cell-suspensions were
washed
and 3x105 cells were resuspended in FACS buffer (PBS containing 0.1% BSA and

53


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
0.1% sodium azide, Sigma Chemicals; St. Louis, MO). Endothelial cells were
incubated with primary antibodies for 30 min at 4 C. If necessary, cells were
resuspended in FACS buffer and stained with a secondary antibody for 30 min at
4 C. Cells were then washed, fixed in 1% paraformaldehyde, and 104 cells were

analyzed by flow cytometry using a FACScalibur instrument and Ce1lQuest
software (Becton Dickinson, San Diego, CA).

Embedding porcine aortic endothelial cells in a three-dimensional
biocompatible matrix altered the expression of surface molecules. Constitutive
expression of CD58 was significantly reduced in porcine aortic endothelial
cells

io embedded in Gelfoam compared to CD58 expression of porcine aortic
endothelial
cells grown on tissue culture polystyrene plates (-60.4%, p<0.002). There was
also
a significant reduction in upregulation of costimulatory and adhesion
molecules, and
MHC class II on matrix-embedded porcine aortic endothelial cells compared to
porcine aortic endothelial cells grown on polystyrene plates under FACS-
analysis

(CD80: -64.9%, p<0.002; CD86: -65.4%, p<0.001; CD40: -53.8%, p<0.005; ICAM-
1: -68.7%, p<0.001; VCAM-1: -53.9%, p<0.005; E-selectin: -71.8%, p<0.0005; P-
selectin: -79.9%, p<0.0002; MHC II: -78.3%, p<0.0002). There were no
significant
differences in surface expression of MHC class I and CD3 1.

Similarly, embedding human aortic endothelial cells in a three-dimensional
2o biocompatible matrix altered the expression of surface molecules. Human
aortic
endothelial cells grown in a 3D matrix exhibited a significantly reduced
expression
profile of CD58 and showed a significant lack in upregulation of costimulatory
and
adhesion molecules. However, there were no significant differences in ICAM-1,
E-
selectin, MHC I, and CD31 expression levels between human aortic endothelial
cells

54


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
embedded in Gelfoam and human aortic endothelial cells grown on tissue culture
polystyrene plates. Furthermore, there were no significant differences in
constitutive expression of PD-L2 (100%, p=0.73) and in upregulation of PD-L1
(86%, p=0.09).

Thus, embedding endothelial cells in a three-dimensional biocompatible
matrix reduces costiinulatory and adhesion molecules. Matrix embedded porcine
aortic endothelial cells and human aortic endothelial cells exhibited
significantly
lower expression levels of costimulatory and adliesion molecules on activated
endothelial cells.

Expression of CD31, MHC-II, CD58, ICAM-1 and E-selectin was also
analyzed in the implants in vitro by confocal microscopy and and in rats in
vivo by
immunohistochemical analysis. Endothelial cells were seeded on cover slips or
embedded in Gelfoam-matrices. After washing with PBS and fixation with 3%
paraformaldehyde for 20 min (cover slips) or overnight (Gelfoam), endothelial
cells

were blocked with rat serum (Bethyl Laboratories, TX) for 30 min. Before
staining
with antibodies, Gelfoam matrices were cut into 2 mm thick slices. Endothelial
cells
were stained with the appropriate amount of antibodies for 1(cover slips) or 2
hours
(Gelfoam) and analyzed on a Zeiss LSM5 10 Laser scanning confocal microscope.
Staining intensity was quantified with ImageJ software (National Institute of
Health,

Bethesda, MD) and normalized against CD31 expression.

Confocal microscopy revealed reduced expression-levels of CD58, ICAM-1,
E-selectin, and MHC-II on matrix embedded porcine aortic endothelial cells
whereas CD31 expression remained unchanged (p<0.02). Cell-substrate anchoring
had no effect on MHC-I expression but markedly muted the expected upregulation



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
of MHC-II molecules. Porcine aortic endothelial cells embedded in Gelfoam
evoked only a modest proliferation of xenogeneic CD4} T cells in-vitro similar
to
the response seen with blockade of MHC-II binding in free porcine aortic
endothelial cells.

Modulation of the immune response in vivo

Matrix embedded porcine aortic endothelial cells showed a lower stimulation
of the initial event in the recruitment of leukocytes which involve P- and E-
selectin,
and of VCAM-1 which is closely associated with T cell recruitment at sites of
immune inflammation. The full panel of general and species specific
costimulatory

molecules was down regulated by matrix embedding, including the first report
of
endothelial cell-expression and suppression of 4-1BB-ligand. At the same time,
expression and upregulation of PD-L1 and PD-L2, members of the B7-fainily that
act as countervailing iiihibitory molecules, remained intact after matrix
embedding.
These in-vitro findings translated into a significantly muted immune reaction
in rats

after implantation of matrix-embedded porcine aortic endotlielial cells.
The cellular response to implantation was also evaluated
immunohistochemically in six rats from each group on day 28 post implantation.
Five-micrometer paraffin sections were cut and antigen retrieval performed by
microwave heating for 10 minutes in a 0.01 mol/L citrate buffer, pH 6Ø

2o Leukocytes, T and B lymphocytes were identified by an avidin-biotin
peroxidase
complex method. The primary antibodies were mouse anti-rat CD45RO, to identify
leukocytes (Research Diagnostics; 1:50 dilution), mouse anti-rat CD4, to
identify
CD4+-T cells (Pharmingen; 1:10 dilution), and mouse anti-rat CD8, to identify
CD8+-T cells (Pharmingen; 1:50 dilution). Rat spleen was used as a positive

56


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
control, and mouse IgG as negative controls. Primary antibodies were applied
for 1
hour at room temperature, and all sections were counterstained with Mayer's
hematoxylin solution (Sigma). Six nonoverlapping fields (x600) were examined.
The results for each group were averaged.

Embedding endothelial cells in a three-dimensional biocompatible matrix, as
compared to injected free PAE or PAE injected adjacent to a three-dimensional
biocompatible matrix, also reduced the immune response in rats in vivo.
Porcine
aortic endothelial cells embedding in Gelfoam significantly reduced formation
of
porcine aortic endothelial cell-specific IgG in vivo. Serum cytokines (MCP-1,
IL-6,

io TNF-a) rose, peaking five days after implantation, in rats receiving free
porcine .
aortic endothelial cells and injections of porcine aortic endothelial cells
adjacent to
Gelfoam. In contrast, cytokine levels did not increase above background in
animals
with matrix-embedded porcine aortic endothelial cells.

Immunohistological studies revealed evidence of cellular infiltration into and
around the implants/injection site at 28 days. After injection of free porcine
aortic
endothelial cells and injection of porcine aortic endothelial cells adjacent
to
Gelfoam, T cells were abundant within the implant/injection side, whereas
large
numbers of CD45RO positive leukocytes were also found at the periphery of the
graft. In contrast, the tissue surrounding the implant and Gelfoam- porcine
aortic

endothelial cells itself were infiltrated with 4.5 fold fewer leukocytes and
CD4}-T
cells, and 3.3 fold fewer CD8+ T cells than the other cell implantation
groups.
Circulating rat immunoglobulins specific for the implanted porcine aortic

endothelial cells were also measured by flow cytometry. 2x105 porcine aortic
endothelial cells were detached from tissue culture polystyrene plates with
0.25%
57


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
trypsin/0.04% EDTA, pelleted, washed, resuspended in FACS buffer and incubated
with serum from recipient rats for 30 min at 4 C (diluted 1:10 in FACS
buffer).
After washing twice with cold FACS buffer, cells were incubated with FITC-
conjugated anti-rat IgG. Following 30 min incubation at 4 C in the dark, the

samples were again washed twice with cold FACS buffer, fixed in 1%
paraformaldehyde, and 10~ cells were analyzed by flow cytometry using a
FACScalibur instrument and Ce1lQuest software. Rat IL-6 (R&D Systems, MN,
detection limit 21 pg/ml), rat TNF-a (R&D Systems, detection limit <5 pg/ml),
and
rat MCP-1 (Amersham, detection limit <5 pg/ml) serum-concentrations were

quantified by ELISA on days 0, 5, 12, and 28 post implantation. Measurements
were performed at the same time by the same ELISA to avoid variations of assay
conditions.

The levels of immunoglobulins specific for the implanted porcine aortic
endothelial cells in serum of the experimental mice were also measured by flow
cytometry. 2x105 porcine aortic endothelial cells, from the same strain as the

implanted cells, were detached from cell culture plates with 0.25%
trypsin/0.04%
EDTA, pelleted, washed, and resuspended in FACS buffer (PBS, 1% FCS, 0.1%
sodium azide). These cells were then incubated with serum froni recipient mice
for
60 min at 4 C (diluted 1:10 in FACS buffer). After washing twice with FACS

buffer, cells were incubated with FITC-conjugated rat anti-mouse IgGZa
(Southern
biotechnology, AL), IgGi (clone A85-1), or IgM (clone R6-60.2, BD Pharmingen,
CA) respectively. Following 30 min incubation at 4 C in the dark, the samples
were
again washed twice with cold FACS buffer, fixed in 0.25 ml 1%
paraformaldehyde,

58


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
and 104 cells were analyzed by flow cytometry using a FACScalibur instrument
and
Ce1lQuest software.

Embedded endothelial cells in a three-dimensional biocompatible matrix, as
compared to injected free PAE and PAE injected adjacent to a three-dimensional

biocompatible matrix, reduced the Th2-driven immune response in mice in vivo.
To
characterize the magnitude and nature of the porcine aortic endothelial cell-
specific
antibody response, serum was collected from mice after implantation of porcine
aortic endothelial cells in the subcutaneous dorsal space as Gelfoam-embedded
cells,
saline-suspended cell pellets, or as pellets adjacent to empty Gelfoam. Post-

1o implantation anti-porcine aortic endothelial cell IgGi and IgM levels were
similar
and significantly higher in mice receiving porcine aortic endothelial cell
pellets or
porcine aortic endothelial cell pellets adjacent to empty Gelfoam compared to
recipients of porcine aortic endothelial cells embedded in Gelfoam (Figures 1A
and

- 1B). There was a transient and minor elevation in anti-porcine aortic
endothelial
cell IgG2a 12 days after implantation (p<0.005) after implantation of matrix-
embedded porcine aortic endothelial cell mice which was not seen in mice
receiving
pelleted porcine aortic endothelial cells or implants of empty Gelfoam with
injection
of pelleted porcine aortic endotlielial cells (Figure 1 C).

Figures 1 A, 1 B and 1 C graphically depict circulating PAE-specific IgG in
mice after subcutaneous injection of free PAE, of Gelfoam-grown endothelial
cells,
or after concomitant injection of PAE adjacent to Gelfoam alone as determined
via
flow-cytometry. Graphic depiction of results from all mice (n=18 per group to
day
28, n=12 per group day 56-100 post-implantation) demonstrates a statistically

significant difference between the matrix-embedded and other forms of PAE
59


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
implantation for IgGi (Figure 1A) and IgM (Figure 1B). There was a transient
and
minor elevation in anti-PAE IgG2a 12 days after implantation of matrix-
embedded
PAE (Figure 1C).

Compared to unstimulated HAE grown on tissue culture plates, matrix-
embedded HAE expressed significantly higher levels of the inhibitory signaling
molecules suppressor-of-cytokine-signaling (SOCS)3 (0.007 0.001 vs.

0.003 0.0003 RU, p<0.001). Hence stimulation with IFN-y resulted in
significantly
lower expression of MHC II on matrix-embedded HAE (374-5 vs. 68 4%, p<0.001).
Despite unchanged IFN-y-receptor expression levels (p=0.39) substrate
adherence

1o reduced IFN-y-induced phosphorylation of Janus kinase 1 and 2 and signal-
transducer-and-activator-of-transcription-1. This was followed by diminished
expression of interferon-regulatory factor-l, CIITA (0.01 0.004 vs.
0.03:L0.004 RU,
p<0.005), and HLA-DR (0.17+0.04 vs. 0.27- 0.05 RU, p<0.02) in matrix-embedded
HAE. Reduced MHC II expression on matrix-embedded HAE resulted in muted

ability to induce proliferation of allogeneic T cells (4152 255 vs. 19619-+327
cpm,
p<0.001).

Interestingly, embedding endothelial cells in a three dimensional matrix
nearly completely diminishes the observed Th2-driven immune response, mutes
lytic activity and attenuates differentiation of naive T cells into effector
cells. In

accordance witli previous results, these data suggest that Gelfoam embedding
of
cells provides immune protection by immune activation at the T-cell level via
reduced expression levels of MHC class II molecules as well as costimulatory
and
adhesion molecules.



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Modulation of lymphocyte proliferation and lytic activity.

Porcine aortic endothelial cells grown on polystyrene wells or embedded in
Gelfoain were seeded in 96 well plates at 5x104 cells/well and stimulated with
40
ng/ml porcine INF-y for 48 hours, followed by mitomycin C treatment (Sigma, 50

g/ml for 30 min) to prevent background proliferation. Human CD4+ lymphocytes
were purified by negative selection with a CD4+ T cell isolation kit II
(Miltenyi
Biotec, Germany) according to the manufacturer's instructions and added at
2x105
cells/well. In some experiments a murine antibody directed against HLA-DP, DQ,
DR blocked activation via MHC class II molecules. 3[H]-thymidine incorporation

was measured on day 6 by 16h pulse (1 Ci/ml, Amersham). Thymidine uptake of
mitoinycin-treated porcine aortic endothelial cells, medium or T cells alone
was
used as negative controls.

To evaluate lymphocyte lytic activity in mice in vivo, splenocyte isolation
and evaluation was performed. Spleens of 4 mice from each group were isolated
aseptically in a laminar flow hood on day 28 after porcine aortic endothelial
cell-

implantation. Organs were cut in several pieces. Clumps were further dispersed
by
drawing and expelling the suspension several times through a sterile syringe
with a
19-Gauge needle. Afterwards, the suspension was expelled through a 200 m mesh
nylon screen. Cells were washed twice with RPMI (containing 2 mM L-glutamine,

0.1 M HEPES, 200 U/ml Penicillin G, 200 g/mi streptomycin and 5% heat-
inactivated calf serum, Life Technologies) and immediately used.

61


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
To further evaluate lymphocyte lytic activity in mice in vivo, a Calcein-AM
release assay was performed. Porcine aortic endothelial cells from the same
strain
of injected cells were resuspended in complete medium at a final concentration
of
2x104/well and incubated with 15 M calcein-AM (Molecular Probes) for 40 min
at
37 C with occasional agitation. After two washes with complete medium,

splenocytes as effector cells were added at a final concentration of
5x105/well.
Spontaneous and maximum release were examined as controls in six replicate
wells
that contained only target cells in coinplete medium and six wells with target
cells in
medium plus 2% Triton X-100 for the last 20 minutes. After 3 hour incubation
at

37 C/5% CO2 samples were measured using a Fluoroskan Ascent FL dual-scanning
microplate luminofluorimeter (Thermo Electron Corporation, TX). Data were
expressed as arbitrary fluorescent units (AFU). Specific lysis was calculated
according to the formula [(test release-spontaneous release)/(maximum release-
spontaneous release)] x 100.

Embedding endothelial cells in a tliree-dimensional biocompatible matrix
reduced lymphocyte proliferation. The proliferative response of isolated human
CD4+ T cells to untreated and INF-y treated porcine aortic endothelial cells
(40
ng/ml. 48 hours) grown in tissue culture plates or embedded in Gelfoam was
assayed by thymidine incorporation. The strong CD4+ T cell proliferation noted

after exposure to porcine aortic endothelial cells pretreated with INF-y was
nearly
eliminated when porcine aortic endothelial cells were matrix-embedded
(17087.2:L3749.75 vs. 5367.8+1976.3 cpm, p<0.01). The presence of MHC II
antibody blocked lymphocyte proliferation in response to INF-y-treated porcine
aortic endothelial cells by 65% to a level comparable to matrix embedded
porcine

62


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
aortic endothelial cells. Mitomycin-treated porcine aortic endothelial cells
did not
show a significant proliferation after 6 day culture (61E 13 cpm) as well as
culture of
isolated CD4+ T cells alone (83 27 cpm).

Similarly, embedding endothelial cells in a three-dimensional biocompatible
matrix, as compared to injected free PAE and PAE injected adjacent to a three-
dimensional biocompatible matrix, reduced lymphocyte lytic activity in mice in
vivo. Lymphocytes from mice spleens from the three different treatment groups
were isolated 28 days after porcine aortic endothelial cell implantation.
Donor
porcine aortic endothelial cells were labeled with Calcein-AM and endothelial
cell-

lysis was measured by a calcein fluorescence release assay after coincubation
with
lymphocytes. Lymphocytes from mice after pure porcine aortic endothelial cell-
injection (36.8f3.9%) and after concomitant porcine aortic endothelial cell-
injection
(33.92:4.7%) showed the highest lytic activity as compared to lymphocytes
isolated
from mice after implantation of porcine aor-tic endothelial cell-Gelfoam
constructs
(22.4 4.2%, p<0.05; Figure 2).

Figure 2 graphically depicts splenocytes from mice receiving free PAE and
shows significantly increased lytic activity when compared to matrix-embedded
PAE. Rechallenge of mice with free PAE significantly increased xenogeneic
lytic
activity of isolated splenocytes.

2o Modulation of Th2 cytokine-producing cells and cytokines:

Immunospot plates (Millipore, Bedford, MA) were coated wit115 g/ml of
anti-mouse interferon (IFN)-y, anti-mouse interleukin (IL)-2, anti-mouse IL-4,
or
anti-mouse IL-10 mAb (all BD Pharmingen) in sterile PBS overnight. The plates
63


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
were then blocked for two hours with complete RPMI-medium without phenol red,
containing 10% heat-inactivated calf serum. Splenocytes (0.5x106 in 100 l
complete RPMI-medium) and the same strain of porcine aortic endothelial cells
used
for implantation (0.5x106 in 100 l complete RPMI-medium) were then placed in

each well and cultured for 48 hours at 37 C in 5% CO2. After washing with
deionized water followed by washing with PBS containing 0.05% Tween (PBST), 2
g/ml of biotinylated anti-mouse IFN-y, anti-mouse IL-2, anti-mouse IL-4, or
anti-
mouse IL-l0 mAb (all BD Pharmingen) were added overnight respectively. The
plates were then washed three times in PBST, followed by one hour of
incubation

with horseradish peroxidase-conjugated streptavidin (BD Pharmingen). After
washing four times with PBST followed by PBS, the plates were developed using
3-
amino-9-ethyl-carbazole (BD Pharmingen). The resulting spots were counted on a
computer-assisted enzyme-linked immunospot image analyzer (Cellular Technology
Ltd., ORT). The number of spots in the wells with medium, splenocytes or
porcine

aortic endotllelial cells alone was subtracted from xenoresponses to account
for
background in data analysis.

Embedding endothelial cells in a three-dimensional biocompatible matrix, as
compared to injected free PAE and PAE injected adjacent to a three-dimensional
biocompatible matrix, reduced Th2 cytokine-producing cells in mice in vivo.
The

frequency of Thl cytokine (IFN-y, IL-2) and Th2 cytokine (IL-4, IL-10)-
producing
cells was measured by ELISPOT assay in splenocytes recovered from animals
after
implantation of different forms of porcine aortic endothelial cells. At day 28
postimplantation, the frequency of Th2 cytokine-producing cells was
significantly
lower in splenocytes isolated from mice receiving matrix embedded porcine
aortic

64


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
endothelial cells compared with those isolated from mice receiving fiee
porcine
aortic endothelial cells or porcine aortic endothelial cells adjacent to empty
Gelfoam
((IL-4: p<0.0001, IL-10 <0.001; Figure 3A). In contrast, there were no
significant
differences in the frequency of Thl cytokine-producing cells in splenocytes
isolated
from the three groups (Figure 3B).

Figure 3A graphically depicts the frequencies of xenoantigen-specific
cytokine-producing cells in recipients after implantation of mice with free
PAE,
matrix-embedded PAE, or PAE injection adjacent to empty Gelfoam. There were
no significant differences in frequency of xenoreactive INF-y and IL-2
producing T-

cells between the three groups on day 28. However, rechallenge with matrix-
embedded PAE evokes a significant increase in xenoantigen-specific INF-7 and
IL-2
producing T-cells.

Figure 3B depicts representative ELISPOT wells for one mouse of each
treatment group 28 days after first implantation and second implantation

respectively. IL-4 production in response to PAE was measured. The number of
IL-4 spots in each well was determined by computer-assisted image analysis.
Figure 3C graphically depicts that recipients of free PAE exhibited a

significant increased frequency of xenoreactive IL-4 and IL-10 producing T-
cells
compared to recipients of matrix-embedded PAE on day 28. Rechallenge with free
PAE or PAE injection adjacent to empty Gelfoam matrices significantly
increased

frequency of xenoantigen-specific IL-4 and IL-10 producing T-cells on day 128.


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Modulation of effector cells.

Splenocytes recovered from the recipients were resuspended in FACS buffer
at a concentration of 2x106/ml. Cells were stained with anti-CD4 FITC (clone
L3T4), antiCD8 FITC (clone Ly-2), anti-CD44 R-PE (clone Ly-24), and anti-

CD62L allophycocyanin (clone Ly-22), and isotype controls (all BD PharMingen).
CD4+ and CD8+ effector cells expressing CD44h'gh and CD62LI W were enumerated,
as previously described.

Embedding endothelial cells in a three-dimensional biocompatible matrix
prevented xenorej ection in mice in vivo. To determine the effect of matrix

embedding on the generation and function of xenoreactive CD4} and CD8+ T
cells,
we measured the number of CD62LI WCD44h'gh found in the spleens of mice
treated
after implantation of matrix-embedded porcine aortic endothelial cells,
implantation
of saline-suspended cell pellets, or as pellets adjacent to empty Gelfoam 28
days
following implantation (Figure 4). CD4+ and 8+ effector cells have been
reliably

identified as CD62L1owCD44nign cells. The percentage of CD62L1ovi'CD44h'gh
cells
increased significantly in free porcine aortic endothelial cell-recipients and
mice
receiving porcine aortic endothelial cells adjacent to empty Gelfoanl compared
with
mice receiving matrix-embedded porcine aortic endothelial cells; the frequency
of
CD4+CD62L' WCD44h'gh T cells outnumbered CD8+ effector cells in all groups
(ratio
1.7-2.3).

Figure 4A graphically plots significantly increased CD4+ and CD8+ effector
cells in mice receiving free PAE. CD4+ effector cells outnumbered CD8+ T-cells
on
days 28 and 128. CD4+ splenocytes recovered from mice were analyzed by flow
cytometry using CD62L and CD44 as markers for effector T cells. Representative
66


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
plots from mice receiving free (a), matrix-embedded (b), or PAE adjacent to
Gelfoam (c) 28 days after implantation.

Figure 4B graphically depicts expansion of effector cells increases after
rechallenge in mice receiving free PAE but not matrix-embedded PAE.

Modulation of xenoreiection and immunological memory.

Embedding endothelial cells in a three-dimensional biocompatible matrix
produced immunological memory after implantation of non-vascularized
xenogeneic
tissue. Thl cytokines play critical roles in the prevention of xenorejection
by down-
regulating the Th2-driven humoral responses. In this regard, the data
demonstrate

i.o that tissue engineered endothelial cells can evoke a significant increase
of porcine
aortic endothelial cell-specific IgG2a antibodies and a significant increase
in
xenoreactive Thl producing splenocytes after rechallenge.

One hundred days after the first implantation, the remaining mice in each
group were rechallenged with porcine aortic endothelial cells identical to
their first
encounter. Mice receiving saline-suspended cell pellets or pellets adjacent to
empty

Gelfoam showed a significant IgGI-driven porcine aortic endothelial cell-
specific
antibody response exceeding the response observed after the first course of
iinplantation (Figure 5A). Only a weak IgM-antibody release was seen (Figure
5B).
In marked contrast, mice receiving matrix-embedded porcine aortic endothelial
cells

did not show an increase in porcine aortic endothelial cell-specific anti-IgGI
and
IgM levels but exhibited a significant release of porcine aortic endothelial
cell-
specific IgGZa antibodies that was absent in the other two mice groups (Figure
5C).

67


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Figures 5A, 5B and 5C graphically depict rechallenge mice (n=12 per group
to day 128, n=6 per group day 156-190 post-implantation) with free PAE or PAE
adjacent to Gelfoam significantly increased formation of PAE-specific IgGi-
antibodies compared to rechallenge with matrix-embedded PAE (Figure 5A).

Rechallenge has no influence on PAE-specific IgM-formation (Figure 5B) and
there
were no significant differences of PAE-specific IgG2a-antibodies between the
three
groups (Figure 5C).

In line with these results, isolated splenocytes from mice receiving free
porcine aortic endothelial cells or porcine aortic endothelial cell-injections
adjacent
to empty Gelfoam showed significantly increased capability to lyse porcine
aortic

endothelial cells 28 days after rechallenge, whereas lysing-capability of
splenocytes
from mice receiving a second implant of matrix-embedded porcine aortic
endothelial cells was significantly weaker than after the first implantation
(Figure 6).
The frequency of xenoreactive IL-4 and IL-10 producing T cells increased

significantly in mice after reimplantation of free porcine aortic endothelial
cells, the
frequency of Th2 producing splenocytes after rechallenge with matrix-einbedded
porcine aortic endothelial cells was unchanged. However, rechallenge with
matrix-
embedded porcine aortic endotlZelial cells induced a higher frequency of
xenoreactive INF-y and IL-2 producing splenocytes than after the first course
of

implantation.

Figure 6 graphically depicts matrix embedding or MHC II blockade restore
proliferation of mice splenocytes exposed to PAE to unstimulated levels. Mice
splenocytes proliferate in response to INF-7 stimulated PAE. Matrix-embedding
68


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
endothelial cells or presence of MHC II antibody blocked splenocyte
proliferation in
response to INF-y treated PAE by -79%. Each value represents mean SD.

Furthermore, 28 days after rechallenge the percentage of CD4+ effector cells
further increased in mice receiving free porcine aortic endothelial cells and

increased significantly in mice receiving porcine aortic endothelial cells
adjacent to
empty Gelfoam implants but remained unchanged in mice receiving matrix
embedded porcine aortic endothelial cells. The same pattern was obvious for
CD8+
effector T cells.

In vitro stimulation of naive mice splenocytes with PAE revealed a

io significantly muted proliferative response of splenocytes when incubated
with INF-y
stimulated matrix-embedded endothelial cells compared to free endothelial
cells.
The presence of MHC II antibody blocked splenocyte proliferation in response
to
INF-y-treated PAE by 79% to a level comparable to matrix embedded PAE.

Overall, the spleen size in mice receiving matrix-embedded porcine aortic
endothelial cells appeared smaller than in the other groups at the end of the
study
period (62.9 9.6, 112.7 16.9, 102.5 18.8 mm3; p<0.05).

Thus, cognate interactions between naive T cells and resting endothelial cells
can lead to tolerance in vitro and in vivo. These data document formation of
immunological memory after implantation of non-vascularized xenogeneic tissue.

Immunological memory was characterized by a significant increase in antigen-
specific IgGi and IgM levels, lytic activity of splenocytes and tendency
towards
increased differentiation into effector T cells. In contrast, rechallenging
mice with
matrix embedding of endothelial cells led to a reduced lytic ability of
splenocytes,

69


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
frequency of effector CD4+ and CD8+ T cells was unchanged. Whereas rechallenge
with matrix-embedded porcine aortic endothelial cells had no influence on
generation of anti-PAE IgGI and IgM, IgG2a levels increased significantly.
Modulation of fractalkine expression.

Chemokines and adhesion molecules are critical in recruiting circulating
immune cells into the vessel wall. Fractalkine has both chemoattractive and
adhesive functions and is involved in the pathogenesis of atherosclerosis,
cardiac
allograft rejection, glomerulonephritis, and rheumatoid arthritis. We compared
expression and secretion of fractalkine between free and matrix-embedded human

aortic endothelial cells (HAE) via RT-PCR, Western blot, flow-cytometry and
ELISA. Adliesion assays were conducted with cytokine-stimulated HAE and 51Cr
labeled natural killer (NK) cells.

HAE were stimulated with 100 U TNFa/ml (Sigma) and 100 U IFN-y/ml
(Roche) at 37 C in a humidified air atmosphere containing 5% C02, conditions
demonstrated to result in maximal fractalkine levels in cultured endothelial
cells.

Flow Cytometry: Endothelial cell monolayers or endothelial cells matrix-
embedded in Gelfoam were harvested after stimulation with TNFa and IFN-y for
indicated time periods. Media were aspirated and cells were washed with PBS.
Monolayers were incubated in 1 mM PBS/EDTA for 5 min, and disrupted by gentle

shaking. Gelfoam-grown cells were digested with collagenase type I
(Worthington
Biochemical, NJ), which was shown to have no effect on CX3CL1-expression.
Cell-suspensions were washed and 3x105 cells fixed in 4% paraformaldehyde for
10
min. After two washing steps, cells were resuspended in saponin-buffer (0.1%



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
saponin, 0.05% NaN3 in Hanks' Balanced Salt Solution), centrifuged and the
supernatant decanted. HAE were then incubated with FITC-conjugated mouse anti-
human CX3CLI (IgGI, clone 51637, R&D Systems, Minneapolis, MN) or a
matched isotype control (clone MOPC-31 C, Pharmingen) for 45 min at 4 C. Cells

were then waslied and 104 cells were analyzed by flow cytometry using a
FACScalibur instrument and Ce1lQuest software.

Western blot analysis: Cell monolayers or cells digested from Gelfoam
matrices by collagenase-treatment were washed in PBS buffer and cell lysates
were
prepared by incubation with lysis buffer (20 mM Tris, 150 mM NaCI, pH 7.5, 1%

Triton X-100, 1% deoxycholate, 0.1% SDS and protease inllibitor; Roche).
Samples
were separated on 4-20% Ready Tris-HCl gels (BioRad Laboratories, Hercules,
CA). A positive control for fractalkine detection was used, consisting of an
85- to
90-kDa form of recombinant human fractalkine lacking the carboxy-termina157
amino acids (R&D Systems). Proteins were then transferred onto PVDF membranes

(Millipore, Billerica, MA) by using glycin-Tris transfer buffer. Blot
membranes
were blocked in Starting Block blocking buffer (Pierce, Rockford, IL) for 1
hour.
For fractalkine-detection, blocked membranes were incubated witll goat anti-
human
fractalkine polyclonal antibody (R&D Systems) at a dilution of 1:200 in
blocking
buffer overnight at 4 C. Membranes were then washed three times at room

temperature with wash buffer consisting of PBS with 0.05% Tween 20 and then
incubated with secondary antibody, a rabbit anti-goat IgG conjugated to
horseradish
peroxidase (Santa Cruz Biotechnology, Santa Cruz, CA) at a 1:3.000 dilution in
blocking buffer for 2 hours at room temperature followed by washing in five
changes of wash buffer. For detection of fractalkine bands, the blot was
incubated

71


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
with chemiluminescence substrate (Western Lightning Chemiluminescence Reagent
Plus kit, Perkin-Elmer, Boston, MA) according to the manufacturer's
instructions
followed by exposure to X-ray film (Kodak X-Omat Blue XB-1).

ELISA: Conditioned mediuin from endothelial cell monolayers or

endothelial cells embedded in Gelfoam after cytokine stimulation was harvested
for
indicated time periods. Secreted fractalkine was detected with a commercially
available enzyme-linked immunosorbent assay (ELISA) detection kit (R&D
Systems). Briefly 96-well Immulon plates (Fisher Scientific, Pittsburgh, PA)
were
coated overnight at room temperature with 100 l of 4 g/ml of mouse anti-
human

fractalkine capture antibody in PBS. After three washes with wash buffer (PBS-
0.05% Tween-20) plates were blocked for 3 h in 1% bovine serum albumin-5%
sucrose in PBS. 100 1 of standards (420 ng/ml of recombinant human
fractalkine
(provided with kit) was used diluted as twofold serial dilutions in diluent
buffer) or
conditioned medium were added, followed by incubation overnight at room

temperature. After three washing steps the plate was incubated with 100 l of
500
ng/ml mouse anti-human fractalkine detection antibody in PBS for 2 hours at
room
temperature followed by incubation with 100 l of streptavidin conjugated to
horseradish-peroxidase for 30 min at room temperature. Color was then
developed
by adding 100 l hydrogen peroxide solution mixed with tetramethylbenzidine

(R&D Systems). The optical density was then read at a wavelength of 450 mn.
NK cell-endothelial cell binding assays: HAE were grown to confluence in
6-well plates (6x105 cell/well) or embedded in Gelfoam matrices and activated
with
100 U TNFa/ml and 100 U IFN-y/ml for 20 hours at 37 C in a h*idified air

atmosphere containing 5% COZ and washed once with PBS. Gelfoam matrices were
72


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
digested with collagenase type I, cells counted and plated at a concentration
of 6x10$
cells/well in 6-well plates for 1 hour to allow adherence. Isolated NK cells
were
incubated with 10 Ci of S1Cr/106 NK cells, washed in PBS and then resuspended
(5
105/well) in 400 l of medium alone or medium containing anti-CX3CR1 antibody

at 20 g/ml for 20 min. The NK cell suspension was added to the endothelial
monolayer under gentle rocking conditions (10 cycles/min). After 30 min the
medium was decanted and the wells were gently washed. Adherent cells were
lysed
by treating with 1% Triton in PBS. Total binding was determined by measuring
individual well-associated counts per minutes using a gamma counter. The
analyses

illustrated were representative of at least three independent experiments.
Matrix-embedding repressed induction of fractalkine mRNA. Whereas
resting endothelial cells grown on tissue culture polystyrene plates or within
a three-
dimensional matrix did not express fractalkine, stimulation of HAE grown on
tissue
culture polystyrene plates with TNFa and IFN-y induced fractalkine mRNA

expression in a time dependent manner. Fractalkine mRNA in HAE grown on tissue
culture polystyrene plates expression peaked at 12 hours stimulation with
cells still
expressing significant amounts of mRNA after stimulation for 24 hours. In
contrast,
induction of fractalkine mRNA expression was significantly reduced in matrix-
embedded endothelial cells at all time points studied. The maxiinum was also

reached after 12 hours cytokine stimulation but was only - 10% of expression
levels
in endothelial cells grown to confluence on tissue culture polystyrene plates
(p<0.0001).

73


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Matrix-embedding inhibited fractalkine protein expression in HAE. Western
blot analysis revealed lower protein expression levels of fractalkine in HAE
embedded within Gelfoam matrices compared to endothelial cells grown on tissue
culture polystyrene plates. There was no fractalkine-specific protein band
detectable

in unstimulated endothelial cells and in endothelial cells stimulated for 4
hours.
Endothelial cells grown on tissue culture polystyrene plates expressed
fractalkine
after 8 hours of stimulation and exhibited maximal expression from 16 to 24
hours
of stimulation with TNFa and IFN-y. Protein-expression in matrix-embedded HAE
was detectable later (12 hours), weaker and disappeared within 24 hours of
cytokine
lo stimulation.

In analogy to Western blot results, flow cytometry analysis revealed
significant higher fractalkine protein expression level in HAE grown on tissue
culture polystyrene plates. Whereas maximal expression on matrix-embedded
endothelial cells was reached after 16 hours of cytokine stimulation (22.8
5.7%),

endothelial cells grown on tissue culture polystyrene plates reached a maximal
and
significant increased fractalkine expression after 20 hours stimulation with
TNFa
and IFN-y (76.5 8.6%; p<0.0001).

Experimental data indicate a reduced secretion of fractalkine from cytokine
stimulated matrix-embedded endothelial cells. Fractalkine levels were also

measured as cumulative levels of soluble fractalkine released into the
endothelial
culture supernatants by ELISA. Levels of soluble fractalkine paralleled those
in
Western blot and flow cytometry analysis: fractalkine secreted from HAE grown
on
tissue culture polystyrene plates significantly exceeded levels secreted by
matrix-
embedded HAE (32.2 2.4 vs. 13.8 1.7 pg/ml after 24 hours of culture;
p<0.0002).

74


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Experimental data indicate a reduced adhesion of NK cells to matrix-
embedded endothelial cells. To study the functional relevance of our finding,
an
adhesion assay with 51Cr labeled NK cell and cytokine-stimulated HAE grown on
tissue culture polystyrene plates or matrix-embedded was performed next. As

revealed by gamma-counting, significantly more NK-cells adhered to allogeneic
HAE grown on tissue culture polystyrene plates than embedded within Gelfoam
(6335 420 vs. 1735 135 cpm; p<0.0002; Fig. 5). The importance of fractalkine
expression for NK cell adhesion to activated endothelial cells could be
demonstrated
as addition of 20 g/ml anti-CX3CL1 significantly augmented adhesion of NK
cells

io. to cytokine stimulated HAE by -74 /o (p<0.005 vs. without anti-CX3CL1). NK
cells did not adhere to tissue culture polystyrene plates or Gelfoam alone.
Modulation of the immune response in heightened immune reactivity mice.'

Endothelial cell injections induced antibody formation in mice. In naive B6
mice three serial subcutaneous injections of 5x105 PAE raised circulating
endothelial
cell-specific IgGI (2210 341 vs. 53 12 mean fluorescence intensity (MFI);

p<0.0001) and IgM antibodies compared to saline injections (136 39 vs. 49 14
MFI; p<0.02). There were no PAE-specific IgG2a antibodies detectable in serum
of
either mouse groups (data not shown) 42 days after first injection of PAE.

Matrix-embedded endothelial cells prevented humoral immune reactivity.
Implantation of matrix-embedded xenogeneic endothelial cells, in marked
contrast
to implantation of free cells, failed to induce a significant humoral immune
response
in naive mice (d 42, IgGi: 210 102 vs. 735 327 MFI; p<0.001; IgM: 60 11 vs.
299 51 MFI; p<0.001; Figures 7A and 7B). Injection of free PAE in pre-
sensitized'



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
serially challenged mice resulted in an elevated humoral immune response with
a
pronounced increase in IgGI antibody-levels (3795 448 MFI; p<0.0002 vs. naive
mice) and slight increase in PAE-specific IgM (164 28 MFI). In marked
contrast,
implantation of matrix-embedded PAE in pre-sensitized serially challenged mice
did

not increase PAE-specific antibodies: moreover antibody-levels specific for
the
injected PAE slowly decreased with time (IgGi: 1578 334 MFI; p<0.0005 vs.
free.
PAE; IgM: 69 5 MFI; p<0.01 vs. free PAE; Figures 7A and 7B). There was no
increase in PAE-specific IgG2a-antibodies in the four treatment groups (data
not
shown) supporting previous reports of a dominating Th2 response in
xenografting.

Figures 7A and 7B graphically depict circulating PAE-specific IgGI (Figure
7A) and IgM (Figure 7B) in naive and pre-sensitized serially challenged mice
after
subcutaneous implantation of non-embedded or matrix-embedded PAE. Graphic
depiction of results from all mice (n=12/group to day 70, n=6/group day 71-132
post-implantation) demonstrates significant differences between matrix-
embedded

and free PAE implantation. Antibody-levels after implantation of matrix-
embedded
PAE slowly diminish. Data are expressed as mean values SD.

Matrix-embedded endothelial cells are poor inducers of cellular immunity.
ELISPOT-analysis revealed a high frequency of xenogeneic T-helper cell (Th)2-
cytokine (IL-4, IL-10) producing splenocytes in naive and pre-sensitized
serially

challenged mice 90 days after implantation of free but not after implantation
of
matrix-embedded PAE. The frequency of xenoreactive splenocytes in pre-
sensitized
serially challenged mice exceeded xenoreactive splenocyte activation and
differentiation in naive mice receiving free PAE (IL-4: 907 59 vs. 680 129;
p<0.02; IL-10: 1096 94 vs. 888 151 number of spots; p<0.02; Figures 8A and
8B).

76


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Yet, compared to implantation of matrix-embedded PAE in naive mice,
implantation
of matrix-embedded PAE in pre-sensitized serially challenged mice elicited
only a
slight increase in IL-4 (322 75 vs. 199 99 number of spots; p<0.05; p<0.0005
vs.
free PAE; Figure 8A) but not in IL-10 producing xenoreactive splenocytes (403
142

vs. 451 135 number of spots; p=0.27; p<0.001 vs. free PAE; Figure 8B).
Significantly fewer Th2-ctyokine producing splenocytes were present in pre-
sensitized serially challenged mice receiving matrix-embedded PAE compared to
naive mice receiving free PAE (p<0.001). The frequency of Thl-cytokine (IFN-7
and IL-2) producing splenocytes did not differ significantly between the four

lo treatment groups again supporting a predominant Th2-role in xenoreactivity
(data
not shown).

Figures 8A and 8B graphically depict the frequencies of xenoreactive
cytokine-producing cells in recipients after implantation of free PAE or
matrix-
embedded PAE in naive and pre-sensitized serially challenged mice. Data are

expressed as mean values SD. Naive and pre-sensitized serially challenged
recipients of free PAE exhibited significant increased frequencies of IL-4
(Figure
8A) and IL-10 (Figure 8B) producing splenocytes compared to recipients of
matrix-
embedded PAE.

The increase in cytokine-producing splenocytes in mice receiving non-

embedded PAE was paralleled by an increase of CD4+ and CD 8+ effector T cells
over time (CD4+: 44 2 naive mice, 54 4% pre-sensitized mice, p<0.05; CD8}: '
20 2; 21 2%; Figures 9A and 9B). Accordingly, differentiation of T cells into
CD44'igh/CD62L1o' T cells was significantly muted in naive and pre-sensitized
77


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
serially challenged mice exposed to matrix-embedded PAE (CD4+: 22 2 naive
mice, 21 3% pre-sensitized mice; p<0.01 vs. free PAE; CD8+: 12 2; 14 3%;
p<0.02 vs. free PAE; Figures 9A and 9B). CD4+ outnumbered CD8+ effector T
cells

1.7-2.6 in all treatment groups. A strong correlation was noted between the

frequency of Th2-cytokine producing splenocytes and extent of T cell
differentiation
cells into CD4+CD44h'9l1/CD62L'0W (IL-4: r=0.81; p<0.0001; IL-10 r=0.88;
p<0.0001; Figure 10) and CD8+CD44h'gl'/CD62L1ow effector cells (IL-4: r=0.79;
p<0.0001; IL-10 r=0.86; p<0.0001) across all treatment groups on day 132.

Figures 9A and 9B graphically depict significantly increased CD4+ (Figure
9A) and CD8+ (Figure 9B) effector cells in mice receiving free PAE.
Splenocytes
recovered from mice were analyzed by flow-cytometry using CD62L and CD44 as
markers for effector T cells. No difference between naive and pre-sensitized
serially
challenged mice when endothelial cells are matrix-embedded. Data are expressed
as
mean values SD.

Figures l0A and lOB are Spearman correlations of the frequencies of Th2-
cytokine producing splenocytes and the extent of T cell differentiation into
effector
cells. Figure 10A graphically depicts the frequency of IL-2 cytokines. Figure
l OB
graphically depicts the frequency of IL-10 cytokines. The correlations suggest
that
cytokine levels correlate linearly with effector T cell induction. Area of the
density
ellipse represents the 95% confidence interval.

Matrix-embedded endothelial cells are shielded from lytic damage. The
ability of host lymphocytes to damage xenogeneic endothelial cells was
characterized on day 70 and day 132. Calcein release plateaued at effector:
target
78


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
ratios of 25:1. For this ratio, endothelial cell damage was 1.6 fold higher in
naive
mice and 1.7 fold higher in pre-sensitized mice when receiving non-embedded in
place of matrix-embedded PAE on d70 (p<0.001). These ratios increased to 1.9
and
2.3 respectively after 132 days (p<0.0005; Figure 11). Of note, the extent of

endothelial damage in pre-sensitized mice receiving matrix-embedded PAE was
significant lower when compared to naYve mice receiving free PAE (20.9 2.3 vs.
37.1 3.4% AFU; p<0.001; Figure 11).

The ability of host lymphocytes to damage xenogeneic endothelial cells was
characterized on day 70 and day 132. Figure 11 graphically depicts the degree
of
damage to endothelial cells in naive and pre-sensitized mice when the
endothelial

cells are free or matrix embedded. Endotlielial damage via lysis is
significantly
reduced in naive and pre-sensitized mice receiving matrix-embedded compared to
free PAE. 2x104 PAE were labeled with calcein and incubated with 5x105
splenocytes isolated after 70 and 132 days respectively.

Calcein release plateaued at effector: target ratios of 25:1. For this ratio,
endothelial cell damage was 1.6 fold higher in naive mice and 1.7 fold higher
in pre-
sensitized mice when receiving non-embedded in place of matrix-embedded PAE on
day 70 (p<0.001). These ratios increased to 1.9 and 2.3 respectively after 132
days
(p<0.0005; Figure 11). Of note, the extent of endothelial damage in pre-
sensitized

mice receiving matrix-embedded PAE was significant lower when compared to
nazve mice receiving free PAE (20.9:L2.3 vs. 37.1:0.4% AFU; p<0.001; Figure
11).
79


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Modulation of dendritic cell maturation.

Dendritic cells are antigen-presented cells that have the unique ability to
both
initiate and regulate immune responses. Mature dendritic cells promote T cell
differentiation into effector and memory cells whereas immature dendritic
cells

present (self-)antigens in a tolerogenic fashion. Dendritic cells are
implicated in a
variety of endothelial-mediated diseases, and activated endothelial cells
induce their
maturation. Because dendritic cells are critical in immune reactivity, it
follows that
endothelial cell-driven dendritic cell maturation is dependent on endothelial
cell-
matrix contact.

ro Preparation, culture and maturation of dendritic cells: Peripheral blood
was
collected froni healthy volunteers and fractionated over Ficoll-Paque (Sigma
Chemicals, St. Louis, MO) by a staildard procedure. To derive dendriti cells,
total
peripheral blood monocytic cells (PBMC) were cultured at 2 x 106 cells/ml in
complete media (RPMI 1640, 10% heat-inactivated calf serum, 0.1 mM sodium

pyruvate (Life Technologies)) for 1.5 hours in tissue culture flasks.
Following
incubation, nonadherent cells were removed by extensive washing with a 1
xsolution
of HBSS (Life Technologies). The remaining adherent cells were then cultured
in
complete media containing 20 ng/ml interleukin (IL)-4 and 20 ng/ml GM-CSF
(Peprotech, Rocky Hill, NJ) for 5 days in a CO2 incubator at 37 C. The
resulting

cells were semi- to nonadherent and MHC IIlOW/CD14'/1ow/CD83' (data not
shown).
For further maturation, adherent and nonadherent dendritic cells were
harvested, extensively washed, counted and 5x105 dendritic cells were
stimulated
with a cytokine cocktail (10 ng/ml IL-I(3, 1000 U/nil IL-6, 20 ng/ml IL-4, GM-
CSF,
and TNF-a; all Preprotech), 1.5x105 HAE or 1.5x105 PAE for 48 hours.
Endothelial



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
cells were either presented as suspensions after grown to confluence on tissue
culture plates or surface adherent embedded within Gelfoam matrices. Every
assay
was repeated at least four times. After maturation, dendritic cells were
isolated from
any contaminating endothelial cells with magnetic bead-labeled CD 1 a
antibodies

(Miltenyi, Bergisch-Gladbach, Germany). Flow cytometry analysis revealed 98%
purity of the isolated DC (data not shown).

Real-time PCR: Total RNA was extracted from isolated dendritic cells and
the remaining endothelial cells using the RNeasy Mini Kit (Qiagen, Valencia,
CA)
according to the manufacturer's instructions. Complementary DNA was
synthesized

using TaqMan reverse transcription reagents from Applied Biosystems (Foster
City,
CA). Real-time PCR analysis was performed with an Opticon Real Time PCR
Machine (MJ Research, Waltham, MA) using SYBR Green PCR Master Mix
(Applied Biosystems) and selected primers. Data from the reaction were
collected
and analyzed by the complementary Opticon computer software. Relative

quantification of gene expression were calculated with standard curves and
normalized to GAPDH.

Flow cytometry: Dendritic cell or endothelial cell suspensions were washed
and 3x105 cells were resuspended in FACS buffer (PBS containing 0,1% BSA and
0,1% sodiuin azide; Sigma Chemicals). Standard flow cytometric analysis
assessed
surface expression of various markers. The following monoclonal antibodies

directly conjugated with phycoerythrin (PE) or fluorescein isothiocyanate
(FITC)
were used in single-color flow cytometric analysis: PE-CDIa (clone HI149,
IgGI),
FITC-CD3 (clone UCHT1, IgGi), PE-CD14 (clone TUK4, IgG2a), PE-CD31 (clone
WM59, IgGI), FITC-CD40 (clone 5C3, IgGi), FITC-CD54 (clone 15.2, IgGI),

81


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
FITC-CD80 (clone BB1, IgM), FITC-CD83 (clone HB15e, IgGI), FITC-CD86
(clone 2331, IgGI), FITC-CD106 (clone 51-10C9, IgGI), FITC-HLA-DP,DQ,DR
(clone CR3/43, IgGI), FITC-Toll-like receptor (TLR)2 (clone TL2.3, IgG2a), and
FITC-TLR4 (clone HTA125, IgG2a). Appropriate isotype control antibodies (mouse

PE-IgGI, PE-IgG2a, FITC-IgGI, FITC-IgGZa, FITC-IgM) were used respectively.
Antibodies were purchased from DakoCytomation (Carpinteria, CA), Serotec
(Raleigh, NC) or PharMingen (San Diego, CA). After staining, cells were washed
and fixed in 1% paraformaldehyde before analysis on a FACScalibur instrument
and
CellQuest software (Becton Dickinson, Mountain View, CA).

Endocytic activity: Endocytic activity of dendritic cells was measured by
uptake of FITC-conjugated dextran (MW 40.000; Molecular Probes, Eugene, OR) as
previously described. Briefly, dendritic cells at various states of maturation
were
incubated in complete media with 1 ingfinl FITC-conjugated dextran for 1 hour
at
37 C to measure specific uptake, or at 4 C to measure nonspecific binding.
Cells

were then washed extensively and analyzed by flow cytometry as described
above.
Mixed lymphocyte reaction assay: CD3+ T-cells from an unrelated donor
were prepared from total PBMC by negative selection using antibody depletion
and
magnetic beads according to the manufacturer's instruction (Dynal Biotech,
Lake
Success, NY). The noiunagnetic fraction contained greater than 95% CD3+ T-
cells,

as assessed by flow cytometry. 2x105 CD3} T-cells/well were seeded in 96-well
round-bottom plates. Purified cytokine- or endothelial cell-matured dendritic
cells
were y-irradiated (3000 rad from a 137Cs source) and added to T-cells at
1x104,
4x103, or 2x103 cells/well to give final ratios of 1:20, 1:50, or 1:100 DC:T-
cells. On
day 5, 1 Ci of 3H-thymidine (Perkin-Elmer, Boston, MA) was added to each
well.

82


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Cells were harvested 18 hours later and 3H-thymidine uptake quantified using a
Packard TopCount y-counter (GMI, Ramsey MI).

Western Blot: After separation from dendritic cells, endothelial cells were
washed in PBS buffer and cell lysates were prepared by incubation with lysis
buffer
(20 mM Tris, 150 mM NaC1, pH 7.5, 1% Triton X-100, 1% deoxycholate, 0.1%

SDS and protease inhibitor; Roche, Indianapolis, IN). Samples were separated
on 4-
20% Ready Tris-HC1 gels (BioRad Laboratories, Hercules, CA). Proteins were
then
transferred onto PVDF membranes (Millipore, Billerica, MA) using glycin-Tris
transfer buffer. Jurkat (TLR2) or HL-60 whole cell lysates (TLR4, both Santa
Cruz

Biotechnologies, Santa Cruz, CA) served as controls. Membranes were blocked in
Starting Block blocking buffer (Pierce, Rockford, IL) for 1 hour. Blocked
membranes were incubated with rabbit anti-human TLR2 (dilution 1:250 in
blocking
buffer) or TLR4 antibodies (dilution 1:200, both Santa Cruz Biotechnologies)
overnight at 4 C. Membranes were then washed three times at room temperature

with wash buffer consisting of PBS with 0.05% Tween 20 and then incubated with
secondary antibody, a goat anti-rabbit IgG conjugated to horseradish
peroxidase
(Santa Cruz Biotechnology, Santa Cruz, CA) at a 1:1.000 dilution in blocking
buffer
for 2 hours at room temperature followed by washing in five changes of wash
buffer.
For detection of TLR bands, the blot was incubated with chemiluminescence

substrate (Western Lightning Chemiluminescence Reagent Plus kit; Perkin-Elmer)
according to the manufacturer's instructions followed by exposure and analysis
on a
FluorChem SP (Alpha Innotech, San Leandro, CA).

83


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Non-adherent endothelial cells directed maturation of monocyte-derived
dendritic cells. In line with previous observations, monocytes differentiated
into
immature dendritic cells after 5 days of culture in GM-CSF and IL-4 (data not
shown). Prolonged cytokine-stimulation with IL-1(3, TNF-a, and IL-6 for 48
hours

upregulated costimulatory (CD40: 2.3 fold compared to immature dendritic
cells,
CD80: 1.9 fold, CD86: 1.6 fold) and HLA-DR molecules (1.5 fold) together with
expression of CD83 (2.2 fold) as an established dendritic cells-maturation
marker.
Exposure to saline suspensions of allo- and xenogeneic endothelial cells after
growth
to confluence in tissue culture plates induced full maturation of monocyte-
derived

dendritic cells to a similar degree as prolonged treatment with a cytokine
cocktail.
HAE or PAE alone induced dendritic cell costimulatory molecule expression with
increases in CD40 (HAE: 2.1 fold, PAE: 2.5 fold compared to immature dendritic
cells), CD80 (2.1 fold, 2.3 fold; p<0.05 vs. cytokine-stimulation), CD86 (1.6
fold,
1.7), HLA-DR (1.7 fold, 2.2 fold; p<0.05 vs. HAE, p<0.002 vs. cytokine-

stimulation), and CD83 (2.6 fold; p<0.05 vs. cytokine stimulation, 3.2 fold;
p<0.02
vs. HAE, p<0.001 vs. cytokine-stimulation).

In a similar fashion, dendritic cell TLR2 and 4 expression were upregulated
upon exposure to saline suspensions of HAE (1.5 and 2.5 fold respectively) to
a
similar or greater extent than cytokine stimulation (1.5 fold for both TLR
compared

to immature dendritic cells). This effect was even more pronounced after co-
incubation of dendritic cells with non-adherent xenogeneic PAE (TLR2: 2.4
fold;
p<0.05 vs. cytokine- and HAE-stimulated, TLR4: 3.0 fold; p<0.05 vs. HAE,
p<0.001 vs. cytokine-stimulation). Similar results could be obtained for mRNA
transcript levels. Additionally, dendritic cells matured with cytokines or non-


84


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
adherent endothelial cells displayed significant upregulation of IL 12 p40
mRNA
(immature: 0.03 0.02 relative units (RU), cytokine-stimulated: 0.23 0.03 RU,
p<0.002, HAE-stimulated: 0.31 0.05 RU, p<0.001, PAE-stimulated: 0.28 0.03,
p<0.002).

Incubation witli substrate-adherent endothelial cells resulted in incomplete
dendritic cell-maturation and sustains endocytic activity. In marked contrast
to co-
culture with non-adherent endothelial cells, co-culture of dendritic cells
with

substrate-adherent HAE and PAE embedded within a three-dimensional matrix
restricted dendritic cell maturation: these dendritic cells displayed only
weak

upregulation of CD40 (substrate-adherent HAE: 1.5 fold compared to immature
DC,
p<0.02 vs. non-adherent HAE, substrate-adherent PAE: 1.3 fold, p<0.002 vs. non-

adherent PAE), CD80 (substrate-adherent HAE and PAE: 1.3 fold, p<0.005 vs. non-

adherent EC), CD86 (substrate-adherent HAE: 1.1 fold, PAE: 1.2 fold, both
p<0.005
vs. non-adherent EC), CD83 (substrate-adherent HAE: 1.5 fold, p<0.001 vs. non-

adlierent HAE, PAE: 1.4 fold, p<0.0002 vs. non-adherent PAE), and TLR4
I
(substrate-adherent HAE: 1.5 fold, PAE: 1.3 fold, both p<0.005 vs. non-
adherent
endothelial cells). Co-incubation with substrate-adherent endothelial cells
failed to
induce HLA-DR and TLR2 expression on dendritic cells at all (p<0.005).
Incubation with empty Gelfoam matrices alone had no effect on maturation of

monocyte-derived dendritic cells (data not shown). Real-time PCR analysis
revealed the same pattern of incomplete maturation when dendritic cells were
exposed to substrate-adherent allo- and xenogeneic endothelial cells.
Induction of
IL12 p40 was similarly significantly weaker when dendritic cells had been
matured



CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
with substrate-adherent endothelial cells (HAE-stimulated: 0.06 0.01, p<0.005,
PAE-stimulated 0.07 0.02, p<0.02).

Immature dendritic cells efficiently captured antigen and exhibited a high
level
of endocytosis. FITC-conjugated dextran uptake increased when monocytes were

cultured for 3 and 5 days in GM-CSF and IL-4 (423.3 121.8 mean fluorescence
intensity (MFI), 239.8 42.8 MFI, p<0.0001). Maturation is typically
accompanied
by concomitant increase in antigen presenting function and reduced capacity
for
antigen capture via endocytic activity. Dextran uptake typically decreases
with
continued cytokine-stimulation (89.7 14.7 MFI, p<0.0001 vs. d5) and with co-

incubation with non-adherent HAE (92 20.3 MFI) or PAE (82.4 16.5 MFI). In
marked contrast, dendritic cells retained their endocytic activity when
endotllelial
cells were presented in a substrate-adherent three-dimensional state and
dextran
uptake was markedly increased (substrate-adherent HAE: 203.2 11.3 MFI, p<0.05
vs. d5, p<0.0001 vs. non-adherent HAE; substrate-adherent PAE: 254.3 32 MFI,
p<0.0001 vs. non-adherent PAE).

Dendritic cells exhibited reduced T-cell proliferation activity after
cultivation
with substrate-adherent endothelial cells. The ability to promote T cell
differentiation into effector and memory cells is an important functional
marker for
the maturation grade of dendritic cells. Whereas cytokine-treated and non-
adherent

endothelial cell exposed dendritic cells induced T-cell proliferation over the
full
spectrum of dendritic cell:T-cell ratios tested (74789 1777, HAE: 97522 1630,
and
PAE: 101616 4302 cpm) this ability was significantly muted in dendritic cells
co-
86


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
incubated with substrate-adherent HAE (18320 1000 cpm, p<0.002) and PAE
(20080 683 cpm, p<0.0001).

Activation of substrate-adherent endothelial cells was reduced when co-
cultured with dendritic cells. Real-time PC, flow-cytometry and Western blot

analysis revealed reduced activation of HAE and PAE after co-culture for 2
days
with dendritic cells. After magnetic-bead based isolation of dendritic cells,
the
remaining cells were greater than 95% pure for the endothelial-cell specific
marker
CD31 (data not shown). Real-time PCR analysis demonstrated reduced mRNA
expression levels for adhesion molecules, CD58, HLA-DR and TLR-molecules on

io substrate-adherent HAE when compared to their non-adherent counterparts.
Reduced mRNA-expression levels translated into reduced surface and
intracellular
expression with 3.6 fold lower expression of 1CAM-1 on substrate-adherent when
compared to non-adherent HAE (1.3 fold decrease for PAE), 4.9 fold decrease of
VCAM-1 for HAE (PAE: 2.7 fold), and 16 fold decrease of HLA-DR for HAE

(PAE: 23 fold decrease). Densitometry analysis of Western blots revealed
increased
TLR2 (HAE: 1.5 fold increase, PAE: 1.6 fold increase; p<0.05) and TLR4
expression (HAE: 2.3 fold increase, PAE: 2 fold increase; p<0.01) in non-
adherent
endothelial cells when compared to substrate-adherent endothelial cells after
co-
incubation with dendritic cells for 48 hours.

Thus, whereas non-adherent endothelial cells induced maturation of
monocyte-derived dendritic cells to an extent similar to that seen with a
cytokine-
cocktail, co-incubation with substrate-adherent endothelial cells induced only
minor
upregulation of mRNA transcript and protein levels of adhesion, costimulatory
and
HLA-DR molecules on dendritic cells. Dendritic cells co-incubated with
substrate-

87


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
adherent endothelial cells also lacked upregulation of IL12 mRNA and CD83
expression that serve as direct maturation markers. The immature state of
dendritic
cells after co-cultivation with substrate-adherent endothelial cells was
mirrored by
sustained ability to uptake dextran. Functionally, whereas dendritic cells
exposed to

non-adherent endothelial cells displayed enhanced T-cell stimulatory activity
in
mixed lymphocyte reactions, T-cell proliferation after exposure to substrate-
adherent
endothelial cell-matured dendritic cells was significantly weaker.

Further Experiments: Effects on Immune Response

Treatment of transplantation rejection: A population of normal (not immune
compromised) organ transplant recipients will be identified. The population
will be
divided into three groups, one of which will receive an effective amount of
the
implantable material of the present invention prior to receipt of a transplant
organ.
A second group will receive an effective amount of implantable material of the
present invention coincident witli receipt of a transplant organ. A third
group will

not receive the implantable material of the present invention, but will
receive a
transplant organ. Reduction of and/or amelioration of an immune response
and/or
an inflammatory response will be monitored over time by evaluating the
proliferation of T-cell lymphocytes and B-cell lymphocytes in serum samples
and by
monitoring the duration of transplant organ acceptance. It is expected that

candidates receiving an effective amount of the implantable material of the
present
invention will display a reduction in proliferation of lymphocytes and/or an
increase
in the duration of transplant organ acceptance.

88


CA 02605080 2007-10-15
WO 2006/116357 PCT/US2006/015555
Treatment of autoimmune disease: A population of patients diagnosed with
an autoimmune disease will be identified. The population will be divided into
two
groups, one of which will receive an effective amount of the implantable
material of
the present invention. Reduction of and/or amelioration of an autoimmune
response

and/or an inflammatory response will be monitored over time by evaluating the
proliferation of T-cell lymphocytes and B-cell lymphocytes in serum samples
and by
monitoring the intensity and duration of symptoms associated with the
autoimmune
disease. It is expected that candidates receiving an effective amount of the

implantable material of the present invention will display a reduction in
proliferation
of lymphocytes and/or a reduction in the frequency and/or intensity of
symptoms.

89

Representative Drawing

Sorry, the representative drawing for patent document number 2605080 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2006-04-21
(87) PCT Publication Date 2006-11-02
(85) National Entry 2007-10-15
Examination Requested 2009-06-03
Dead Application 2012-12-03

Abandonment History

Abandonment Date Reason Reinstatement Date
2011-12-05 R30(2) - Failure to Respond

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2007-10-15
Maintenance Fee - Application - New Act 2 2008-04-21 $100.00 2008-04-01
Expired 2019 - The completion of the application $200.00 2008-04-14
Maintenance Fee - Application - New Act 3 2009-04-21 $100.00 2009-04-06
Request for Examination $800.00 2009-06-03
Maintenance Fee - Application - New Act 4 2010-04-21 $100.00 2010-04-07
Maintenance Fee - Application - New Act 5 2011-04-21 $200.00 2011-04-06
Maintenance Fee - Application - New Act 6 2012-04-23 $200.00 2012-04-05
Registration of a document - section 124 $100.00 2013-02-06
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MASSACHUSETTS INSTITUTE OF TECHNOLOGY
SHIRE REGENERATIVE MEDICINE, INC.
Past Owners on Record
EDELMAN, ELAZER R.
METHE, HEIKO
NUGENT, HELEN MARIE
PERVASIS THERAPEUTICS, INC.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Claims 2009-03-25 8 302
Description 2009-03-25 92 4,082
Drawings 2007-10-15 12 392
Claims 2007-10-15 8 218
Abstract 2007-10-15 1 72
Description 2007-10-15 89 4,010
Cover Page 2008-01-23 1 44
PCT 2007-10-15 3 142
Assignment 2007-10-15 4 119
Correspondence 2008-01-21 1 28
Prosecution-Amendment 2009-03-25 15 498
Correspondence 2008-04-14 4 146
Prosecution-Amendment 2009-06-03 2 55
Correspondence 2010-02-25 1 28
Prosecution-Amendment 2011-06-03 4 212
Assignment 2013-02-06 17 985